Abstract
A major drawback of single-cell ATAC-seq (scATAC-seq) is its sparsity,
i.e., open chromatin regions with no reads due to loss of DNA material
during the scATAC-seq protocol. Here, we propose scOpen, a
computational method based on regularized non-negative matrix
factorization for imputing and quantifying the open chromatin status of
regulatory regions from sparse scATAC-seq experiments. We show that
scOpen improves crucial downstream analysis steps of scATAC-seq data as
clustering, visualization, cis-regulatory DNA interactions, and
delineation of regulatory features. We demonstrate the power of scOpen
to dissect regulatory changes in the development of fibrosis in the
kidney. This identifies a role of Runx1 and target genes by promoting
fibroblast to myofibroblast differentiation driving kidney fibrosis.
Subject terms: Gene regulatory networks, Machine learning, Renal
fibrosis
__________________________________________________________________
scATAC-Seq yields data that is extremely sparse. Here, the authors
present a computationally efficient imputation method called scOpen
that improves the downstream analyses of scATAC-Seq data and use it to
identify transcriptional regulators of kidney fibrosis.
Introduction
The simplicity and low cell number requirements of the assay for
transposase-accessible chromatin using sequencing (ATAC-seq)^[40]1 made
it the standard method for detection of open chromatin (OC), enabling
the first study of OC of cancer cohorts^[41]2. Moreover, careful
consideration of digestion events by the enzyme Tn5 allowed insights on
regulatory elements such as positions of nucleosomes^[42]1,[43]3,
transcription factor (TF) binding sites, and the activity level of
TFs^[44]4. The combination of ATAC-seq with single-cell sequencing
(scATAC-seq)^[45]5 further expanded ATAC-seq applications by measuring
the OC status of thousands of single cells from healthy^[46]6,[47]7 and
diseased tissues^[48]8. Computational tasks for analysis of scATAC-seq
include detection of cell types with clustering (scABC^[49]9,
cisTopic^[50]10, SnapATAC^[51]11); identification of TF regulating
individual cells (chromVAR^[52]12); and prediction of co-accessible DNA
regions in groups of cells (Cicero^[53]13).
Usually, the first step for analysis of scATAC-seq data is the
detection of OC regions by calling peaks on the scATAC-seq library by
ignoring cell information. Next, a matrix is built by counting the
number of digestion events per cell in each of the previously detected
regions. This matrix usually has a very high dimension (up to >10^6
regions) and a maximum of two digestion events are expected for a
region per cell. As with scRNA-seq^[54]14–[55]16, scATAC-seq is
affected by dropout events due to the loss of DNA material during
library preparation. These characteristics render the scATAC-seq count
matrix sparse, i.e. 3% of non-zero entries. In contrast, scRNA-seq have
less severe sparsity (>10% of non-zeros) than scATAC-seq due to smaller
dimension (< 20,000 genes for mammalian genomes) and lower dropout
rates for genes with high or moderate expression levels. This sparsity
poses challenges in the identification of cell-specific OC regions and
is likely to affect downstream analysis as clustering and detection of
regulatory features. Although several computational methods have been
developed to address this issue for scRNA-seq data (e.g., MAGIC^[56]14,
scImpute^[57]17, DCA^[58]18, and SAVER^[59]19), these methods were not
designed to deal with the sparse and low count nature of scATAC-seq
data. Until date, there are only two approaches for imputation methods
for scATAC-seq data e.g., SCALE^[60]20 and cisTopic^[61]10. SCALE,
which is based on deep learning, requires a graphics processing unit
(GPU) for training. The usual small size of GPU memory limits the
number of cells to be analyzed. cisTopic is a Bayesian-based method,
which was reported to have an exponential increase of the running time
for an increasing number of reads^[62]21. Therefore, both approaches
are likely to have scalability issues with large data sets.
We here present scOpen, an unsupervised learning model for scATAC-seq
data imputation. It estimates accessibility scores to indicate if a
region is open in a particular cell. scOpen is based on a non-negative
matrix factorization (NMF), which makes no assumption on the data
distribution as SCALE or cisTopic. It also includes a regularization,
which makes it less prone to overfitting. To speed up the learning, we
make use of a cyclic coordinate descent (CCD) algorithm. Moreover, we
adopt an elbow detection approach to automatically determine the number
of dimensions of the input data. The imputed matrix can be used as
input for usual computational methods of scATAC-seq data as clustering,
visualization, and prediction of DNA interactions (Fig. [63]1a). We
demonstrate the power of scOpen on a comprehensive benchmarking
analysis using publicly available scATAC-seq data with true labels.
Moreover, we use scOpen together with HINT-ATAC^[64]4 footprinting
analysis to infer regulatory networks driving the development of
fibrosis with a scATAC-seq time-course dataset of 31,000 cells in
murine kidney fibrosis, identifying Runx1 as a regulator of
myofibroblast differentiation.
Fig. 1. scOpen and benchmarking of scATAC-seq imputation methods.
[65]Fig. 1
[66]Open in a new tab
a scOpen receives as input a sparse peak by cell count matrix. After
matrix binarization, scOpen performs TF–IDF transformation followed by
NMF for dimension reduction and matrix imputation. The imputed or
reduced matrix can then be given as input for scATAC-seq methods for
clustering, visualization, and interpretation of regulatory features. b
Memory requirements of imputation/denoising methods on benchmarking
datasets. The x-axis represents the number of elements of the input
matrix (number of OC regions by cells). c Same as b for running time
requirements. d Boxplot showing the evaluation of imputation/denoising
methods for recovering true peaks. The y-axis indicates the area under
the precision-recall curve (AUPR). Methods are ranked by the mean AUPR.
The asterisk and the two asterisks mean that the method is outperformed
by the top-ranked method (scOpen) with significance levels of 0.05 and
0.01 at a confidence level of 0.95 (Wilcoxon Rank Sum test, paired,
two-sided), respectively (n = 1224 cells for Cell lines, n = 2210 cells
for Hematopoiesis, n = 765 cells for T-cells, and n = 10,032 for PBMC).
The box plot represents the median (central line), first and third
quartiles (box bounds). The whiskers present the 1.5 interquartile
range (IQR) and external dots represent outliers (data greater than or
smaller than 1.5IQR). e Barplots showing silhouette score (y-axis) for
benchmarking datasets. f Barplots showing the clustering accuracy for
distinct imputation methods. The y-axis indicates the mean adjusted
Rand Index (ARI). Dots represent individual ARI values of distinct
clustering methods. Error bars represent the standard deviation (SD) of
ARI. Data are represented as mean ± SD. The asterisk and the two
asterisks mean that the method is outperformed by the top-ranked method
with significance levels of 0.05 and 0.01 at a confidence level of 0.95
(n = 8 independent clustering experiments, Wilcoxon Rank Sum test,
paired, two-sided), respectively. Source data for Fig. 1 are provided
as a Source Data file.
Results
OC estimation with scOpen
scOpen performs imputation and denoising of a scATAC-seq matrix via a
regularized NMF based on a binarized scATAC-seq cell count matrix,
where features represent OC regions which are obtained by peak calling
based on aggregated scATAC-seq profiles. This matrix is transformed
using the term frequency-inverse document frequency (TF-IDF), which
weighs the importance of an OC region to a cell. Next, it applies a
regularized NMF using a coordinate descent algorithm^[67]22. In
addition, it provides a computational approach to optimize the
dataset-specific rank k of the NMF approach based on a knee detection
method^[68]23. scOpen provides as results imputed and reduced dimension
matrices, which can be used for distinct downstream analysis as
visualization, clustering, inference of regulatory players, and
cis-regulatory DNA interactions (Fig. [69]1a).
First, we made use of simulated scATAC-seq similar as in ref. ^[70]21
to evaluate the parameterization of two hyper-parameters of scOpen,
i.e., the rank k and the regularization term λ (see the “Methods”
section; Supplementary Fig. [71]1a–d). Results indicate that the scOpen
automatic procedure for rank selection obtains close to optimal
results, i.e. selected rank had similar accuracy than best ranks for
both imputation and clustering problems. Regarding λ, a value of 1 is
optimal in the imputation problem, where values in the range [0, 1]
were optimal for the clustering problem. This indicates the importance
of the regularization parameter in scATAC-seq data imputation. The
λ = 1 and the rank selection strategy are used as default by scOpen.
Benchmarking of scOpen for imputation of scATAC-seq
For benchmarking, we made use of four public scATAC-seq data sets: cell
lines^[72]5, human hematopoiesis composing of eight cell types^[73]6,
four sub-types of T cells^[74]8, and a multi-omics RNA-ATAC from
peripheral blood mononuclear cells (PBMCs) with 14 cell types (see the
“Methods” section). These datasets were selected due to the presence of
external labels, which were defined independently of the scATAC-seq at
hand. After processing, we generated a count matrix for each dataset
and detected 50k to 120k OC regions with 3–7% of non-zero entries,
confirming the sparsity of scATAC-seq data (Supplementary Table [75]1).
For comparison, we selected top-performing imputation/denoising
methods^[76]24 proposed for scRNA-seq (MAGIC^[77]14, SAVER^[78]19,
scImpute^[79]17, DCA^[80]18, and scBFA^[81]25); two scATAC-seq
imputation methods (cisTopic^[82]10 and SCALE^[83]20); a PCA-based
imputation method (imputePCA^[84]26); and the raw count matrix
(Supplementary Fig. [85]2a).
We first evaluated the time and memory requirement of imputation
methods (see the “Methods” section). scOpen had the overall lowest
memory requirements, i.e it required at least 2 fold less memory as
compared to cisTopic, MAGIC, or SCALE (Fig. [86]1b) and had a maximum
requirement of 16 GB on the PBMC dataset (Supplementary Data [87]1).
Regarding computing time, MAGIC was the fastest followed by SCALE and
scOpen. These were the only methods performing the imputation of the
large PBMCs dataset (10k cells vs. 100k peaks) in < 3 h (Fig. [88]1c),
while imputePCA, SAVER, and DCA failed to execute at the PBMCs dataset.
We next tested if imputation methods can improve the recovery of true
OC regions. For this, we created true and negative OC labels for each
cell type by peak calling of bulk ATAC-seq profiles. Next, we evaluated
the correspondence between imputed scATAC-seq values and peaks of the
corresponding cell type with the area under precision-recall curve
(AUPR) (see the “Methods” section). scOpen significantly outperformed
all competing methods by presenting the highest mean AUPR
(Fig. [89]1d). The combined ranking indicates SCALE and MAGIC as
runner-up methods (Supplementary Fig. [90]2b). Next, we evaluated the
influence on the number of cells per cluster in the AUPR. Despite an
overall decrease in AUPR with sample size, we observed that top
performing methods (scOpen, SCALE, and MAGIC) were less sensible to
cell numbers (Supplementary Fig. [91]2c).
We also investigated the impact of imputation on the estimation of
distances between cells and the impact on standard clustering methods.
Distance between cells was evaluated with the silhouette score, while
clustering accuracy was evaluated with adjusted Rand index (ARI)^[92]27
both regarding the agreement with known cell labels. scOpen was the
best performer in all data sets regarding the silhouette score
(Fig. [93]1e). The combined ranking demonstrated that scOpen had
significantly better results than competing methods, while cisTopic and
MAGIC were runner-up methods (Supplementary Fig. [94]2d). Regarding
clustering, scOpen was best in the hematopoiesis and multi-omics PBMCs
datasets and second-best for cell lines and T cell datasets
(Fig. [95]1f). When considering the combined ranking, scOpen performed
best followed by cisTopic and MAGIC (Supplementary Fig. [96]2e). Visual
representations with UMAP^[97]28 projections of these datasets and
methods are provided in Supplementary Fig. [98]3. Altogether, these
results support that scOpen outperforms state-of-the-art imputation
methods while providing the lowest memory footprint and above-average
time performance.
Benchmarking of scATAC-seq clustering methods
Another relevant question was to compare scOpen with top-performing
state-of-the-art scATAC-seq pipelines: cisTopic, SnapATAC and
Cusanovich2018^[99]21 (see the “Methods” section; Supplementary
Fig. [100]4a). Here, pipelines were evaluated with the default
clustering methods, i.e graph-based clustering for SnapATAC^[101]11 and
density-based clustering for other methods^[102]10. We also evaluated
the use of both reduced and imputed matrices for scOpen and cisTopic,
as these methods provide both types of representations.
The evaluation of distance matrices with the silhouette score indicated
that both imputed or low dimension scOpen matrices presented the
highest score in all data-sets (Fig. [103]2a) and both scOpen matrix
representations tied as first in the combined rank (Supplementary
Fig. [104]4b). cisTopic, which was the runner-up method, performed well
in cell lines, hematopoiesis, and T-cells but poorly for multi-omics
PBMCs. Next, we evaluated the clustering performance of competing
pipelines. Again, scOpen performed best on cell lines and hematopoiesis
data sets and ranked first/second in the combined rank (Supplementary
Fig. [105]4c). Overall, this analysis indicates that both reduced
dimension and imputed scOpen matrices obtain the best overall results
for distance and clustering representations on evaluated datasets. Of
note, the low-dimensional matrix reduces the memory footprint on
clustering by more than 1000 fold in comparison to using full imputed
matrices and serves as an alternative for cluster analysis of large
dimensional data sets.
Fig. 2. Benchmarking of scATAC-seq clustering and downstream analysis.
[106]Fig. 2
[107]Open in a new tab
a Bar plot showing an evaluation of distances estimated on distinct
scATAC-seq representations with a silhouette score. b Bar plots showing
the clustering accuracy (ARI) for distinct clustering pipelines. c
Scatter plot comparing silhouette score of datasets by providing raw
(x-axis) and scOpen estimated matrices (y-axis) as input for Cicero and
chromVAR. Colors represent datasets and shapes represent methods. scABC
is not evaluated as it does not provide a space transformation. d Same
as c for clustering results (ARI) of Cicero, chromVAR, and scABC. e
Precision-recall curves showing the evaluation of the predicted links
on GM12878 cells using the raw and imputed matrix as input. We used
data from pol-II ChIA-PET as true labels. Colors refer to methods. We
reported the AUPR for the top 3 methods. f Same as e by using Hi-C data
as true labels. g Visualization of co-accessibility scores (y-axis) of
Cicero predicted with raw and scOpen estimated matrices contrasted with
scores based on RNA pol-II ChIA-PET (purple) and promoter capture Hi-C
(green) around the CD79A locus (x-axis). For ChIA-PET, the
log-transformed frequencies of each interaction PET cluster represent
co-accessibility scores, while the negative log-transformed p-values
from the CHiCAGO software indicate Hi-C scores. h Scatter plot showing
single-cell accessibility scores estimated by top-performing imputation
methods (according to f) for the link between peak 1 and peak 2
(supported by Hi-C data). Each dot represents a cell and color refers
to density. Pearson correlation is shown on the left-upper corner.
Source data for Fig. 2 are provided as a Source Data file.
Improving scATAC-seq downstream analysis using scOpen estimated matrix
Next, we tested the benefit of using scOpen estimated matrices as input
for scATAC-seq computational pipelines, which have as objective the
identification of regulatory features associated with single cells
(chromVAR^[108]12), estimation of gene activity scores and
DNA-interactions (Cicero^[109]13), or a clustering method tailored for
scATAC-seq data (scABC^[110]9) (Supplementary Fig. [111]4d). Both
chromVAR and Cicero first transform the scATAC-seq matrix to either TFs
and genes feature spaces respectively. Clustering was then performed
using the standard pipelines from each approach. We compared the
clustering accuracy (ARI) and distance (silhouette score) of these
methods with either raw or scOpen estimated matrices. In all
combinations of methods and datasets, we observed a higher or equal
ARI/silhouette whenever a scOpen matrix was provided as input
(Fig. [112]2c, d). Results can be inspected with UMAP visualization
with and without scOpen imputation (Supplementary Fig. [113]5).
Prior to estimating gene-centric OC scores, Cicero first predicts
co-accessible pairs of DNA regions in groups of cells, which
potentially form cis-regulatory interactions. We compared Cicero
predicted interactions on human lymphoblastoid cells (GM12878) by using
Hi-C and ChIA-PET from this cell type as true labels for all imputation
methods with data as provided in ref. ^[114]13. Both AUPR values and
odds ratios indicated that the scOpen matrix improves the detection of
GM12878 interactions globally (Fig. [115]2e, f; Supplementary
Fig. [116]6a, b). To evaluate the impact on the number of cell on these
predictions, we have down-sampled the data to only consider 50% or 25%
of cells. We observed a residual decrease in the AUPR of scOpen for 25%
of cells (Supplementary Fig. [117]6c). This supports that chromatin
conformation prediction works well even for cell types with low
abundance. The power of scOpen imputation was clear when checking the
individual locus (Fig. [118]2g), as previously described by
Cicero^[119]13. This is evident when contrasting accessibility scores
between pairs of peak-to-peak links supported by Hi-C predictions
(Fig. [120]2h; Supplementary Fig. [121]6d–h). scOpen obtained highly
correlated accessibility scores, while other imputation methods showed
quite diverse association patterns. Together, these results indicated
that the use of scOpen estimated matrices improves downstream analysis
of state-of-the-art scATAC-seq methods.
Applying scOpen to scATAC-seq of fibrosis driving cells
Next, we evaluated scOpen in its power to improve the detection of
cells in a complex disease dataset. For this, we performed whole mouse
kidney scATAC-seq in C57Bl6/WT mice in homeostasis (day 0) and at
two-time points after injury with fibrosis: 2 and 10 days after
unilateral ureteral obstruction (UUO)^[122]29,[123]30. Experiments
recovered a total of 30,129 high-quality cells after quality control
with an average of 13,933 fragments per cell, a fraction of reads in
promoters of 0.46, and high reproducibility (R > 0.99) between
biological duplicates (Supplementary Fig. [124]7a, b; Supplementary
Table [125]1). After data aggregation, 150,593 peaks were detected,
resulting in a highly dimensional and sparse scATAC-seq matrix (4.2% of
non-zeros).
Next, we performed data integration for batch effect removal using
Harmony^[126]31. For comparison, we used a dimension reduced matrix
from either LSI (Cusanovich2018), cisTopic, SnapATAC, or scOpen. We
annotated the scATAC-seq profiles using single nuclei RNA-seq
(snRNA-seq) data of the same kidney fibrosis model from an independent
study^[127]32 via label transfer^[128]33 to serve as cell labels. We
then evaluated the batch correction results using silhouette score and
clustering. We observed that clusters based on scOpen were more similar
to the transferred labels (higher ARI) than clusters based on competing
methods (Fig. [129]3a). Furthermore, scOpen also provided better
distance metrics and visualization than competing methods
(Supplementary Figs. [130]7c–e and [131]8). These results support the
discriminative power of scOpen in this large and complex dataset.
Fig. 3. scOpen characterizes the progression of kidney fibrosis.
[132]Fig. 3
[133]Open in a new tab
a ARI values (y-axis) contrasting clustering results and transferred
labels using distinct dimensional reduction methods for scATAC-seq.
Clustering was performed by only considering UUO kidney cells on day 0
(WT), day 2, or day 10 or the integrated data set (all days). b UMAP of
the integrated UUO scATAC-seq after doublet removal with major kidney
cell types: fibroblasts, descending loop of Henle and thin ascending
loop of Henle (DL & TAL); macrophages (Mac), Lymphoid (T and B cells),
endothelial cells (EC), thick ascending loop of Henle (TAL), distal
convoluted tubule (DCT), collecting duct-principal cells (CD-PC),
intercalated cells (IC), podocytes (Pod) and proximal tubule cells (PT
S1; PT S2; PT S3; Injured PT). c Proportion of cells of selected
clusters on either day 0, day 2 or day 10 experiments. d Heatmap with
TF activity score (z-transformed) for TFs (y-axis) and selected
clusters (x-axis). We highlight TFs with the decrease in activity
scores in injured PTs (Rxra and Hnf4a), with high TF activity scores in
injured PTs (Batf:Jun; Smad2:Smad3) and immune cells (Creb1; Nfkb1). e
Transcription factor footprints (average ATAC-seq around predicted
binding sites) of Rxra, Smad2::Smad3 and Nfkb1 for selected cell types.
The logo of underlying sequences is shown below and the number of
binding sites is shown top-left corner. f Transcription factor
footprints of Rxra, Smad2::Smad3, and Nfkb1 for injured PT cells in day
0, day 2, and day 10. Source data for Fig. 3 are provided as a Source
Data file.
Next, we annotated the clusters of scOpen by using known marker genes
and transferred labels after removing doublets with ArchR^[134]34. We
identified all major kidney cell types including PT cells,
distal/connecting tubular cells, collecting duct and loop of Henle,
endothelial cells (ECs), fibroblasts as well as the rare populations of
podocytes and lymphocytes (Fig. [135]3b; Supplementary Fig. [136]9a).
Lymphocytes were not described in the previously scRNA-seq
study^[137]32, which supports the importance of annotation of
scATAC-seq clusters independently of scRNA-seq label transfer. Of
particular interest were cell types with population changes during the
progression of fibrosis (Fig. [138]3c; Supplementary Fig. [139]9b–d).
We observed an overall decrease of normal proximal tubular (PT),
glomerular and ECs and an increase of immune cells as expected in this
fibrosis model with tubule injury, the influx of inflammatory cells,
and capillary loss^[140]35,[141]36. Importantly, we detected an
increased PT sub-population, which we characterized as injured PT by
increased accessibility around the PT injury markers Vcam1 and Kim1
(Havrc1)^[142]37 (Supplementary Fig. [143]9a).
Dissecting cell-specific regulatory changes in fibrosis
Next, we adapted HINT-ATAC^[144]4 to dissect regulatory changes in
scATAC-seq clusters. For each cluster, we created a pseudo-bulk
ATAC-seq library by combining reads from single cells in the cluster.
We then performed footprinting analysis and estimated TF activity
scores for all footprint-supported motifs. We only kept TFs with
changes (high variance) in TF activity scores among clusters. We
focused here on clusters associated with PT cells, fibroblasts, and
immune cells, as these represent key players in kidney remodeling and
fibrosis after injury. As shown in Fig. [145]3d, the TF activity scores
capture regulatory programs associated with these three major cell
populations (Supplementary Data [146]2). Injured PTs have overall lower
TF activity scores than all TFs of the PT cluster. TFs with a high
decrease in activity in injured PTs include Rxra, which is important
for the regulation of calcium homeostasis in tubular cells^[147]38, and
Hnf4a, which is important in PT development^[148]39 (Fig. [149]3d, e).
Footprint profile of Rxra in injured PTs display a gradual loss of TF
activity over time indicating that injured PT acquires a
de-differentiated phenotype during fibrosis progression and tubular
dilatation (Fig. [150]3f). A group of TFs with high activity scores in
injured PTs also have increased TF activity scores in fibroblasts
(Smad2:Smad3 and Batf:Jun) indicating shared regulatory programs in
these cells. Smad proteins are downstream mediators of TGFβ signaling,
which is a known key player of fibroblast to myofibroblast
differentiation and fibrosis^[151]40. The high activity of Smad2:Smad3
also indicates a role of TGFβ in the de-differentiation of injured PTs.
Also, both Smad2:Smad3 reach a peak in TF activity level at day 2 after
UUO in injured PTs (Fig. [152]3f), which indicates these TFs are
activated post-transcriptionally. We also detect the high activity of
Nfkb1 in injured PTs (and lymphocytes), which fits with the known role
of Nfkb1 in injured and failed repair PTs^[153]41,[154]42. Moreover,
our analysis also shows a gradual TF activity increase over time in
injured PT (Fig. [155]3f), suggesting that Nfkb1 plays an important
role in sustaining the injured PT phenotype.
scOpen reveals TF driving myofibroblast differentiation
A key process in kidney injury is fibrosis, which is caused by the
differentiation of fibroblasts and pericytes to matrix secreting
myofibroblasts^[156]43. To dissect potential differentiation
trajectories, we performed a diffusion map embedding of the fibroblasts
(Fig. [157]4a), which revealed the presence of three major branches
formed by fibroblasts, pericytes, and myofibroblasts, as supported by
the accessibility of Scara5, Ng2 (Cspg4), Postn and Col1a1
(Supplementary Fig. [158]10)^[159]43,[160]44.
Fig. 4. Role of Runx1 in myofibroblast differentiation.
[161]Fig. 4
[162]Open in a new tab
a Diffusion map showing sub-clustering of fibroblasts. Colors refer to
sub-cell-types and arrow represents differentiation trajectory from
fibroblast to myofibroblast. Pe pericyte, Fib fibroblast, MF
myofibroblast. b Line plots showing cell proportion from the day after
UUO along the trajectory. c Pseudotime heatmap showing gene activity
(left) and TF motif activity (right) along the trajectory. d
Footprinting profiles of Runx1 and Twist2 binding sites along the
trajectory. e Immuno-fluorescence (IF) staining of Runx1 (red) in
PDGFRb-eGFP mouse kidney. In sham-operated mice, Runx1 staining shows a
reduced intensity in PDGFRb-eGFP+ cells compared to remaining kidney
cells (arrows). f Immuno-fluorescence (IF) staining of Runx1 (red) in
PDGFRb-eGFP mouse kidney at 10 days after UUO as compared to sham.
Arrows indicate Runx1 staining in expanding PDGFRb-eGFP+
myofibroblasts. g Quantification of Runx1 nuclear intensity in
PDGFRb-eGFP+ cells in sham vs. UUO mice. Error bars represent the SD of
the intensity. Data are presented as mean ± SD. Statistical
significance was assessed by a two-tailed Student’s t-test with
p < 0.05 being considered statistically significant (n = 3 mice). h
Performance of top-performing imputation methods on the prediction of
Runx1 target genes measured with AUPR. i Peak-to-Gene links (top)
predicted on scOpen matrix and associated to Tgfbr1 in fibroblast
cells. The height of links represents its significance. Dash line
represents the threshold of significance (FDR = 0.001). ATAC-seq tracks
(below) were generated from pseudo-bulk profiles of
fibroblast/myofibroblast cells with increasing pseudo time (0–20,
20–40, 40–60, 60–80, and 80–100). Binding sites of Runx1 (B1–B4)
supported by ATAC-seq footprints and overlapping to peaks are
highlighted on the bottom. j Scatter plot showing gene activity of
Tgfbr1 and normalized peak accessibility from raw (upper) or scOpen
imputed matrix (lower) for peak-to-gene link B4. Each dot represents
cells in a given pseudotime and the overall correlation is shown in the
left-upper corner. Scale bars in e and f represent 50 μm. For details
on statistics and reproducibility, see the “Methods” section. Source
data for Fig. 4 are provided as a Source Data file.
We next created a cellular trajectory across the differentiation from
fibroblasts to myofibroblasts using ArchR (Fig. [163]4a; Supplementary
Fig. [164]10c). We observed that there is an increase in cells after
injury (Day 2 and Day 10) along the trajectory (Fig. [165]4b). We next
characterized TFs by correlating their gene activity with TF activity
along the trajectory (Fig. [166]4c) and ranked these by their
correlation (Supplementary Fig. [167]10d). The correlation of Runx1,
which has a well-known function in blood cells^[168]45, stood out,
besides showing a steady increase in activity in myofibroblasts.
Another TF with high correlation and similar myofibroblast specific
activity was Twist2, which has a known role in epithelial to
mesenchymal transition in kidney fibrosis^[169]46 (Fig. [170]4d).
To validate the yet uncharacteristic role of Runx1 in myofibroblasts,
we performed immunostaining and quantification of Runx1 signal
intensity in transgenic PDGFRb-eGFP mice that genetically tag
fibroblasts and myofibroblasts^[171]43,[172]47. Runx1 staining in
control mice (sham) revealed positive nuclei in tubular epithelial
cells and rarely in PDGFRb-eGFP+ mesenchymal cells (Fig. [173]4e). In
kidney fibrosis after UUO surgery (day 10), Runx1 staining intensity
increased significantly in PDGFRb+ myofibroblasts (Fig. [174]4f, g).
Next, we performed retroviral overexpression experiments and
RNA-sequencing in a human kidney PDGFRb+ fibroblast cell-line that we
have generated^[175]43 to ask whether Runx1 might be functionally
involved in myofibroblast differentiation in humans (Supplementary
Fig. [176]11a, b). Runx1 overexpression led to reduced proliferation
(Supplementary Fig. [177]11c) and strong gene expression changes
(Supplementary Fig. [178]11d). Gene ontology (GO) and pathway
enrichment analysis indicated enrichment of cell adhesion, cell
differentiation, and TGFB signaling following Runx1 overexpression
(Supplementary Fig. [179]11e). Various extracellular matrix genes (Fn1,
Col13A1), as well as a TGFB receptor (Tgfbr1) and Twist2, were
up-regulated following Runx1 overexpression (Supplementary
Fig. [180]11d). Furthermore, we observed increased expression of the
myofibroblast marker gene Postn after Runx1 overexpression. Altogether,
this suggests that Runx1 might directly drive myofibroblast
differentiation of human kidney fibroblasts since overexpression
reduced cell proliferation and induced expression of various
myofibroblast genes.
Identification of Runx1 target genes
Another important application of scATAC-seq is the prediction of
cis-regulatory DNA interactions (peak-to-gene links) by measuring the
correlation between gene activity and reads counts in proximal peaks.
To compare the impact of imputation on this task, we predicted
peak-to-gene links in fibroblasts on distinct scATAC-seq matrices using
ArchR^[181]34 after imputation with top-performing imputation methods.
The use of imputation methods led to improved signals on peak-to-gene
links predictions as indicated by higher correlation values after
imputation (Supplementary Fig. [182]12a, b). We considered all genes
with at least one link, where the peak has a footprint supported
Runx1-binding site, as Runx1 targets. We then compared the predicted
Runx1 targets from distinct scATAC-seq imputed matrices with
differentially expressed genes after Runx1 over-expression (true
labels). All imputation methods obtained higher AUPR values than the
use of a raw matrix, while scOpen obtained the highest AUPR
(Fig. [183]4h; Supplementary Fig. [184]12c). Among others, scOpen
predicted Tgfbr1 and Twist2 as prominent Runx1 target genes
(Fig. [185]4i; Supplementary Fig. [186]12d). We observed several peaks
with high peak-to-gene correlation, increasing accessibility upon
myofibroblast differentiation and presence of Runx1-binding sites. The
positive impact of imputation was clear when observing scatter plots
contrasting gene activity and peak accessibility of these peak-to-gene
links (Fig. [187]4j; Supplementary Fig. [188]12e–i).
Another interesting question is the association of the predicted link
with distinct regulatory features. While we observed no clear
association of the correlation of predicted links with the size of the
link (Supplementary Fig. [189]13a), our analysis suggested that links
associated with active kidney enhancers have a higher correlation than
other active regulatory regions. This further supports the functional
relevance of predicted links. These results suggest that Runx1 is an
important regulator of myofibroblast differentiation by regulating the
EMT-related TF Twist2 and by amplifying TGFB signaling by increasing
the expression of a TGFB receptor 1 and affecting the expression of
extracellular matrix genes. Altogether, these results uncover a complex
cascade of regulatory events across cells during the progression of
fibrosis and reveal a yet unknown function of Runx1 in myofibroblast
differentiation in kidney fibrosis.
Discussion
In ATAC-seq, Tn5 generates a maximum of 2 fragments per cell in a small
(~200 bp) OC region. Subsequent steps of the ATAC-seq protocol cause
loss of a large proportion of these fragments. For example, only DNA
fragments with the two distinct Tn5 adapters, which are only present in
50% of the fragments, are amplified in the PCR step^[190]48. Further
DNA material losses occur during single-cell isolation, liquid
handling, sequencing, or by simple financial restrictions of sequencing
depth. Assuming that 25% of accessible DNA can be successfully
sequenced, we expect that 56% of accessible chromatin sites will not
have a single digestion event causing the so-called dropout events,
assuming that digestion events follow a binomial distribution. Despite
this major signal loss, imputation and denoising have been widely
ignored in the scATAC-seq
literature^[191]5,[192]6,[193]8,[194]9,[195]12,[196]13 and common
scATAC-seq pipelines (e.g., Signac^[197]49 and ArchR^[198]34).
We demonstrated here that scOpen estimated matrices have a higher
recovery of dropout events and also improved distance and clustering
results when compared to imputation methods for
scRNA-seq^[199]14,[200]17–[201]19,[202]25 and the few available
imputations methods tailored for scATAC-seq (cisTopic-impute^[203]10,
SCALE^[204]20). scOpen also presented very good scalability with the
lowest memory requirements and tractable computational time on large
data sets. From a methodological perspective, scOpen is the only method
performing regularization of estimated models to prevent over-fitting.
This is in line with a previous study, which indicated over-fitting as
one of the largest issues on scRNA-seq imputation^[205]50. Moreover, it
is also possible to use the scOpen factorized matrix as a dimension
reduction. We have shown that both dimensions reduced and imputed
matrices from scOpen displayed the best performance on distance
representation and clustering when compared to diverse state-of-the-art
scATAC-seq dimension reduction/clustering pipelines (cisTopic,
SnapATAC, and Cusanovich et al. 2018). Of note, the ArchR pipeline is
equivalent to Cusanovich et al. 2018 and based on the same dimension
reduction method (LSI). It is worth noting that LIGER^[206]51 is
another method of employing NMF for single-cell ATAC-seq analysis. It
uses integrative NMF to extract shared factors with the objective of
multi-modal data integration of scATAC-seq and scRNA-seq. Moreover,
denoising in bulk ATAC-seq has also been approached with the use of
deep learning methods^[207]52. These closely related approaches,
however, have distinct applications than scOpen and are therefore not
evaluated here.
Finally, we have demonstrated that the use of scOpen-corrected matrices
improves the accuracy of existing state-of-art scATAC-seq methods
(cisTopic^[208]10, chromVAR^[209]12, Cicero^[210]13). Particularly
positive results were obtained in the prediction of chromatin
conformation with Cicero, where all methods perform better than raw
matrices. Cicero works by measuring the correlation between pairs of
proximal links. Due to the fact that dropout events are independent for
two regions, it is not surprising that imputation has strong benefits.
This is equivalent to observations from van Dijk et al. 2018^[211]14 in
the context of scRNA-seq, where the prediction of gene–gene
interactions after MAGIC imputation was significantly improved.
Altogether, these results support the importance of dropout event
correction with scOpen in any computational analysis of scATAC-seq. Of
note, a sparsity similar to scATAC-seq is also expected in single-cell
protocols based on DNA enrichment such as scChIP-seq^[212]53,[213]54,
scCUT&Tag^[214]55, or scBisulfite-seq^[215]56. Denoising and imputation
of count matrices from these protocols represent a future challenge.
Moreover, we used scOpen to characterize complex cascades of regulatory
changes associated with kidney injury and fibrosis. Our analyses
demonstrated that a major expanding population of cells, i.e. injured
PTs, myofibroblasts, and immune cells, share regulatory programs, which
are associated with cell de-/differentiation and proliferation. Of all
methods evaluated, scOpen obtained the best clustering results in the
kidney cell repertoire using a scRNA-seq on the same kidney injury
model as a reference. Trajectory analysis identified Runx1 as the major
TF driving myofibroblast differentiation, which was validated by Runx1
staining in the mouse model and by retroviral over-expression studies
in human PDGFRb+ kidney cells. Computational prediction with
peak-to-gene links combined with footprint-supported Runx1-binding
sites indicated the role of Runx1 in the regulation of Tgfbr1 and
Twist2. These were validated on over-expression experiments in human
fibroblasts. Altogether, results suggest that Runx1 makes fibroblasts
more sensitive to TGFB signaling via increasing expression of the TGFB
receptors.
Runx1 has recently been reported as a potential inducer of EMT in PT
cells^[216]57. Furthermore, in vitro data of mesenchymal stem cells
(MSCs) isolated from bone marrow or prostate gland points towards a
potential myofibroblast differentiation role of Runx1^[217]58. In vivo
evidence for a functional role of Runx1 in regulating fibrogenesis has
been demonstrated in zebrafish^[218]59. Single-cell RNA-seq data from
zebrafish heart after cryo-injury suggests that endocardial cells and
thrombocytes up-regulate Runx1 while Runx1 mutant zebrafish
demonstrated enhanced cardiac regeneration after cryoinjury with an
ameliorated fibrotic response. Here we show for the first time in vivo
and in vitro evidence that Runx1 in myofibroblasts regulates scar
formation following a fibrogenic kidney injury in mice. Runx1
deficiency caused reduced myofibroblast formation and enhanced
recovery. To this end, inhibiting Runx1 could lead to reduced
myofibroblast differentiation and increased endogenous repair after
fibrogenic organ injuries in the kidney and heart. Our results shed
light on mechanisms of myofibroblasts differentiation driving kidney
fibrosis and chronic kidney disease (CKD). Altogether, this
demonstrates how scOpen can be used to dissect complex regulatory
processes by footprinting analysis combined with peak-to-gene link
predictions.
Methods
scOpen
scOpen aims to simultaneously impute and reduce the dimension of a
scATAC-seq matrix. Let
[MATH: X∈Rm×
n :MATH]
be the scATAC-seq matrix, where x[ij] is the number of cutting sites in
peak i and cell j; m is the total number of peaks and n is the number
of cells. We first define a binary open/closed chromatin matrix
[MATH: X^∈{0,1}m×n
msup> :MATH]
, i.e.
[MATH:
xij
′=1xij
mi>>00xij
mi>=0 :MATH]
1
where 1 indicates the peak i is open and 0 indicates closed in cell j.
Next, we calculate a score for peak i and cell j by applying term
frequency-inverse document frequency (TF-IDF) transformation:
[MATH:
xij
″=
xij
′∑p=1mxpj′⋅
logn∑
q=1n
xiq<
/mi>′ :MATH]
2
This score represents how important the peak i is for cell j. Next, we
normalize the TF-IDF matrix as
[MATH:
vij=xij″<
/mrow>∑k=1m
msubsup>xkj″
mrow>2 :MATH]
3
We next impute the matrix by minimization of the following optimization
problem:
[MATH: M^=argmin∑i,j(mij
msub>−vij<
/mrow>)2+λ∣∣M∣∣<
mo>*,s.t.mij≥0 :MATH]
4
where
[MATH: ∣∣M∣∣<
mo>*=∑
ikσi(M) :MATH]
is the nuclear norm of matrix M, and σ[i] denotes the ith largest
singular value of M. The first item is the estimator of square loss for
each element in M and λ is the regularization parameter, which aims to
prevent the model from over-fitting and set to 1 as default value. To
solve this problem, we assume that M is a low-rank matrix with rank k
and it can be written as
[MATH: minW,Hf(W,H)=∑ij((WH)ij−v<
mi>ij)<
mn>2+λ2∣∣W∣∣<
mn>2+λ2∣∣H∣∣<
mn>2,s.t.(WH)ij≥0 :MATH]
5
where
[MATH: W∈Rm×
k,H∈Rk×
n :MATH]
. This constrained optimization problem is solved by using CCD
methods^[219]60. This method iteratively updates the variable w[it] in
W to z by solving the following one-variable sub-problem:
[MATH:
minzf<
/mi>(z)=<
munderover accent="false"
accentunder="false">∑j=1n∑t
mi>′∈kwit′h
mi>t′j−with
tj+z
htj−vij<
/msub>2+<
mfrac>λ2z2,s.t.z≥0 :MATH]
6
Likewise, the elements in H can be updated with a similar update rule.
The above iteration is carried out until a termination criterion is
met, e.g. number of iteration performed. Afterward, we calculate M as
the product of W and H to obtain the scOpen imputed matrix or consider
H as scOpen reduced matrix. This algorithm has a theoretical time
complexity of O((m + n)k) for a single iteration and thus is scalable
for large datasets.
Selection of hyper-parameters in scOpen
There are two hyper-parameters in scOpen, i.e., the rank of the matrix
k and regularization parameter λ. Rank k determines the intrinsic
dimensions of a matrix and thus is highly dataset-specific. To select
an appropriate value of k, we first input a number of ranks and
generated a residual sum of squares (RSS) curve in a pre-defined
interval (2–30 as default). Next, we use a knee point detection method
^[220]23, which finds a k with the best trade-off between fit error and
model complexity. We make use of a simulated scATAC-seq dataset as
described below to evaluate the impact of λ and k in either the
imputation and clustering performance (Supplementary Fig. [221]1). We
use optimal settings (λ = 1) and knee detection from an interval of
2–30 in further results.
scATAC-seq simulation dataset
To generate a simulation scATAC-seq dataset, we downloaded bulk
ATAC-seq data of 13 FACS-sorted human primary blood cell types from
gene expression omnibus (GEO) with accession number
[222]GSE74912^[223]61. For each cell type, we processed the data
similarly as in ref. ^[224]12. First, the downloaded files were
converted to FastQ using the SRA toolkit
([225]http://ncbi.github.io/sra-tools/). Next, adapter sequences and
low-quality ends were trimmed from FastQ files using Trim
Galore^[226]62. Reads were mapped to the genome hg19 using
Bowtie2^[227]63 with the following parameters ( − X
2000 − − very − sensitive − − no − discordant), allowing paired-end
reads of up to 2 kb to align. Then, reads mapped to chrY, mitochondria
and unassembled “random” contigs were removed. Duplicates were also
removed with Picard and reads were further filtered for alignment
quality of > Q30 and required to be properly paired using
SAMtools^[228]64. Peaks were called using MACS2^[229]65 with the
following parameters ( − − keep − dup auto − − call − summits). We next
merged the peaks from all cell types to create a unique peaks list. We
then created a peak cell-type matrix by offsetting +4 bp for forward
strand and −5 bp for the reverse strand to represent the cleavage event
center^[230]1,[231]4 and counting the number of reads start sites per
cell type in each peak. This provides a cell type vs. peak matrix A,
where a[ij] indicates the number of reads for cell j in peak i.
We next used this bulk ATAC-seq counts matrix A to simulate a
scATAC-seq counts matrix X by improving the simulation strategy
proposed in ref. ^[232]21. Specifically, given m peaks and T cell
types, we define the accessibility x[ij] ∈ {0, 1} of a single cell j
from the cell type t in peak i as
[MATH:
xij~Bernoulli(p
it<
mo>)pi
t=rit⋅nj⋅(
1−q)+1m⋅n
j⋅qrit=ait∑k=1makt
mrow>nj=Nj
mrow>⋅fN
j~NB(r,<
mi>p) :MATH]
7
where
[MATH:
pit
:MATH]
denotes the probability of cell type t being accessible in peak i, q is
a noise parameter, n[j] denotes the number of reads in peaks for
single-cell j, f denotes the fraction of reads in peaks (FRiP) and N[j]
denotes the total number of reads for cell j. N[j] is sampled from a
negative binomial distribution, whose parameters were estimated from a
real scATAC-seq dataset. We simulated 200 cells per cell type using
this process and used noise q = 0.6 and FRiP f = 0.3. Our approach
differs from ref. ^[233]21 by sampling the number of reads per cell
from a negative binomial distribution rather than using a fixed number
and the introduction of the FRiP parameters.
scATAC-seq benchmarking datasets
The cell line dataset was obtained by combining single-cell ATAC-seq
data of BJ, H1-ESC, K562, GM12878, TF1, and HL-60 from ref. ^[234]5,
which was downloaded from GEO with accession number [235]GSE65360. The
hematopoiesis dataset includes scATAC-seq experiments of sorted
progenitor cells populations: hematopoietic stem cells (HSC),
multipotent progenitors (MPP), lymphoid-primed multipotent progenitors
(LMPP), common myeloid progenitors (CMP), common lymphoid progenitors
(CLP), granulocyte–macrophage progenitors (GMP),
megakaryocyte–erythroid progenitors (MEP), and plasmacytoid dendritic
cells (pDC)^[236]6. Sequencing libraries were obtained from GEO with
accession number [237]GSE96769. In both datasets, the original cell
types were used as true labels for clustering as in the previous
work^[238]9,[239]10. The T cell dataset is based on human Jurkat T
cells, memory T cells, naive T cells, and Th17 T cells obtained from
[240]GSE107816^[241]8. Labels of memory, naive, and Th17 T cells were
provided in Satpathy et al. ^[242]8 by comparing scATAC-seq profiles
with bulk ATAC-seq of corresponding T cell sub-populations.
For each of these three datasets, we only kept cells with at least 500
unique fragments. We then created a pseudo-bulk ATAC-seq library by
merging the obtained scATAC-seq profiles and called peaks using
MACS2^[243]65. The peaks were extended ± 250 bp from the summits as in
ref. ^[244]1 and peaks overlapping with ENCODE blacklists
([245]http://mitra.stanford.edu/kundaje/akundaje/release/blacklists/hg1
9-human/) were removed. we next constructed a peak by cell counts
matrix. To the test scalability of imputation methods, we also included
a multiome PBMC dataset with 10,000 cells
([246]https://support.10xgenomics.com/single-cell-multiome-atac-gex/dat
asets). This dataset was generated using the Chromium Single Cell
Multiome ATAC + Gene Expression assay. We use here the cell types as
annotated by the 10X Genomics R&D team using only the scRNA-seq data.
See Supplementary Table [247]1 for complete statistics associated with
these data sets.
Comparison between scOpen and competing imputation methods
We compared the performance of scOpen with 8 competing imputation
approaches, i.e., MAGIC^[248]14, SAVER^[249]19, scImpute^[250]17,
DCA^[251]18, cisTopic^[252]10, scBFA^[253]25, SCALE^[254]20, and
imputePCA. We performed imputation with these algorithms (see details
below) on the benchmarking datasets.
We first tested if the imputed matrix can recover the true signal for
every single cell. To this end, we used cell labels from each dataset
to aggregate the ATAC-seq profiles and performed peak calling to find
cell-specific OC regions. OC regions present in a particular cell type
were considered as trues and OC regions not present in that cell type
as negatives. For a particular cell, we can obtain true positives and
true negatives by comparing the labels of the corresponding cell type
with the presence of reads (or openness score) in that OC region and
single cell. We use these statistics to measure the area under the
precision-recall curve (AUPR)^[255]66 for each cell.
Next, we evaluated the imputed matrix using the mean silhouette score
of cells^[256]67. For a given cell x:
[MATH: silhouette(x)=b(<
mrow>x)−a(<
/mo>x)max(a
(x),b(x)) :MATH]
8
where a(s) is the average distance between x and the other cells of the
same class, and b(x) is the average distance between x and cells in the
closest different class. The distance was calculated as a 1−Pearson
correlation. A higher silhouette score indicates a higher similarity of
a cell to cells of the same cell type than cells from other cell types.
We next tested if the imputed matrix improves cell clustering. We
applied PCA (50 PCs) for each of the imputed matrices and clustered
cells using k-medoids and hierarchical clustering methods with
1−Pearson correlation as distance. Besides PCA, we also used
t-SNE^[257]68 embedding as input and euclidean as distance, as this
approach is explored by cisTopic^[258]10. We also tested the different
numbers of clusters, e.g. k and k + 1, where k is the true number of
clusters. We used the ARI to evaluate the clustering results^[259]27
with labels from benchmarking data sets (see Supplementary Fig. [260]1
for experimental design). The ARI measures similarity between two data
clustering results by correcting the chance of grouping elements.
Specifically, given two partitions of a dataset D with n cells,
U = {U[1], U[2], ⋯ U[r]} and V = {V[1], V[2], ⋯ , V[s]}, the number of
common cells for each cluster i and j can be written as
[MATH:
cij=∣Ui<
/msub>∩Vj∣ :MATH]
9
where i ∈ {1, 2, ⋯ , r} and j ∈ {1, 2, ⋯ , s}. The ARI can be
calculated as follows:
[MATH:
ARI=∑ijcij
2−∑iai2∑jbj2
mrow>/n2
mrow>12∑iai2+∑jbj2
mrow>−∑iai2∑jbj2
mrow>/n2
mrow> :MATH]
10
where
[MATH:
ai=∑j=1sc<
mi>ij :MATH]
and
[MATH:
bj=∑ir
mrow>cij<
/mrow> :MATH]
, respectively. The ARI has a maximum value 1 and an expected value of
0, with 1 indicating that the data clustering are exactly same and 0
indicating that the two data clustering agree randomly.
We describe below details of running all competing methods.
MAGIC: MAGIC is an algorithm for alleviating sparsity and noise of
single-cell data using diffusion geometry^[261]14. We downloaded MAGIC
from [262]https://github.com/KrishnaswamyLab/MAGICand applied it to the
count matrix with the default setting. Prior to MAGIC, the input was
normalized by library size and root squared, as suggested by the
authors^[263]14.
SAVER: SAVER is a method that recovers the true expression level of
each gene in each cell by borrowing information across genes and
cells^[264]19. We obtained SAVER from
[265]https://github.com/mohuangx/SAVER and ran it on the normalized tag
count matrix with the default parameters.
scImpute: scImpute is a statistical method to accurately and robustly
impute the dropouts in scRNA-seq data^[266]17. We downloaded scImpute
from [267]https://github.com/Vivianstats/scImpute and executed it using
the default setting except for the number of cell clusters which is
used to determine the candidate neighbors of each cell by scImpute. We
defined this as the true cluster number for each benchmarking dataset.
DCA: DCA is a deep auto-encoder network for denoising scRNA-seq data by
taking the count structure, over-dispersed nature, and sparsity of the
data into account^[268]18. We obtained DCA from
[269]https://github.com/theislab/dca and ran it with the default
setting.
cisTopic-impute: cisTopic is a probabilistic model to simultaneously
identify cell states (topic-cell distribution) and cis-regulatory
topics (region-topic distribution) from single-cell epigenomics
data^[270]10. We downloaded it from
[271]https://github.com/aertslab/cisTopic and ran it with different
numbers of topics (from 5 to 50). The optimal number of topics was
selected based on the highest log-likelihood as suggested in ref.
^[272]10. We then multiplied the topic-cell and the region-topic
distributions to obtain the predictive distribution^[273]10, which
describes the probability of each region in each cell and is used as
the imputed matrix for clustering and visualization. We termed this
method cisTopic-impute.
scBFA: scBFA is a detection-based model to remove technical variation
for both scRNA-seq and scATAC-seq by analyzing feature detection
patterns alone and ignoring feature quantification
measurements^[274]25. We obtained scBFA from
[275]https://github.com/quon-titative-biology/scBFA and ran it on the
raw count matrix using default parameters.
SCALE: SCALE combines the variational auto-encoder (VAE) and the
Gaussian mixture model (GMM) to model the distribution of
high-dimensional sparse scATAC-seq data^[276]20. We downloaded it from
[277]https://github.com/jsxlei/SCALE and ran it with the default
setting. We used option impute to get the imputed data.
imputePCA: We also included the principal component-based imputation
method (termed here as imputePCA) on incomplete data sets as a control
for comparison. This method is based on an interactive and regularized
PCA algorithm to predict missing entries, which are considered latent
variables.^[278]26. We installed R package missMDA and performed
imputation with function imputePCA with default settings. All zero
entries were considered as missing data.
Evaluation of time and memory requirement of imputation methods
To compare the memory and running time requirement of each imputation
method, we ran all of them on a dedicated HPC node with the same
computation resources quota, i.e., 180 GB memory, 120 h time, and 4
CPUs. For DCA and SCALE, two deep learning-based methods, we used GPU
with 16 GB memory. We measured the max memory usage during the running
of a method and observed that all methods but imputePCA, SAVER, and DCA
can successfully generate the imputed matrix for all datasets
(Fig. [279]1b). For the multi-omics PBMC dataset, DCA failed due to a
GPU memory issue and we could not obtain results from imputePCA and
SAVER after 120 h of running.
Comparison between scOpen and competing dimension reduction methods
We next compared the performance of scOpen with cisTopic^[280]10,
SnapATAC^[281]11, and latent semantic indexing (LSI) (termed here as
Cusanovich2018)^[282]69 for dimension reduction of scATAC-seq data. We
applied these methods to obtain a low-dimension matrix from each
dataset (detailed below) and measured the mean silhouette score^[283]67
(see Fig. [284]1e).
cisTopic: We downloaded it from
[285]https://github.com/aertslab/cisTopic and ran it with different
numbers of topics (from 5 to 50). The optimal number of topics was
selected based on the highest log-likelihood as suggested in ref.
^[286]10. The topic-cell distribution is used as a dimension-reduced
matrix.
SnapATAC: SnapATAC is a software package for analyzing scATAC-seq
datasets^[287]11. Instead of using peak annotation as features, it
resolves cellular heterogeneity by directly comparing the similarity in
genome-wide accessibility profiles between cells. Furthermore, SnapATAC
uses the Nyström method to generate a low-rank embedding for a
large-scale dataset which enables the analysis of scATAC-seq up to a
million cells. We installed SnapATAC from
[288]https://github.com/r3fang/SnapATAC and followed the tutorial from
[289]https://github.com/r3fang/SnapATAC/blob/master/examples/10X_brain_
5k/README.md to perform dimension reduction for benchmarking datasets.
Cusanovich2018: Cusanovich2018 first segments the genome into 5kb
windows and then scored each cell for any insertions in these windows,
generating a large, sparse, binary matrix of 5 kb windows by cells.
Based on this matrix, the top 20,000 most commonly used sites were
retained. Then, the matrix was normalized and re-scaled using the term
frequency-inverse document frequency (TF-IDF) transformation. Next,
singular value decomposition (SVD) was performed to generate a
PCs-by-cells low dimension matrix.
Benchmarking of scATAC-seq downstream analysis methods
Next, we compared the performance of state-of-art scATAC-seq methods
(scABC, chromVAR, and Cicero) when presented with either scOpen imputed
or raw scATAC-seq matrix. The rationale is if we improve the count
matrix by imputation, we should be able to improve downstream analysis.
Note that scABC is the only method providing a clustering solution.
chromVAR and Cicero transform the scATAC-seq matrices into TF and gene
space. We here again evaluated the results based on clustering accuracy
with methods used as the standard by these pipelines, i.e.,
hierarchical clustering with complete agglomeration method for chromVAR
and k-medoids for Cicero. Moreover, we evaluated the co-accessible
links predicted by Cicero between using scOpen imputed or raw counts
matrix.
scABC: scABC is an unsupervised clustering algorithm for single-cell
epigenomic data^[290]9. We downloaded it from
[291]https://github.com/SUwonglab/scABC and executed it according to
the tutorial
[292]https://github.com/SUwonglab/scABC/blob/master/vignettes/Clusterin
gWithCountsMatrix.html.
chromVAR: chromVAR is an R package for analyzing sparse
chromatin-accessibility data by measuring the gain or loss of chromatin
accessibility within sets of genomic features, as regions with sequence
predicted TF-binding sites^[293]12. We obtained chromVAR from
[294]https://github.com/GreenleafLab/chromVAR and executed to find
gain/loss of chromatin accessibility in regions with binding sites of
571 TF motifs obtained in JASPAR version 2018^[295]70.
Cicero: Cicero is a method that predicts co-accessible pairs of DNA
elements using single-cell chromatin accessibility data^[296]13.
Moreover, Cicero provides a gene activity score for each cell and gene
by assessing the overall accessibility of a promoter and its associated
distal sites. This matrix was used for clustering and visualization of
scATAC-seq. We obtained Cicero from
[297]https://github.com/cole-trapnell-lab/cicero-release and executed
it according to the document provided by
[298]https://cole-trapnell-lab.github.io/cicero-release/docs/.
Chromosomal conformation experiments with Cicero
We used conformation data as true labels to evaluate co-accessible
pairs of cis-regulatory DNA as detected by Cicero on GM12878 cells. We
obtained a scATAC-seq matrix of GM12878 cells from GEO
([299]GSM2970932). For evaluation, we downloaded promoter-capture (PC)
Hi-C data of GM12878 from GEO ([300]GSE81503), which uses the
CHiCAGO^[301]71 score as a physical proximity indicator. We also
downloaded ChIA-PET data of GM12878 from GEO ([302]GSM1872887), which
used the frequency of each interaction PET cluster to represent how
strong the interaction is. We considered all obtained links, as
provided by these data sets, as true interactions as in ref. ^[303]13.
To investigate the performance of each method against the number of
cells, we also randomly down-sampled the data to 50% and 25%. Next, we
replicated the evaluation analysis performed in Fig. 4 of ref. ^[304]13
and contrasted the results of Cicero with raw or matrices obtained
after scOpen imputation. Next, we use the built-in function
compare_connections of Cicero to define the true labels for predicted
co-accessibility links. Using the correlation as prediction, we finally
computed the area of precision and recall curve (AUPR) with pr.curve
function from R package PRROC^[305]72.
Unilateral ureter obstruction (UUO) mouse kidney animal experiments
UUO was performed as previously described^[306]30. Shortly, the left
ureter was tied off at the level of the lower pole with two 7.0 ties
(Ethicon) after flank incision. One C57BL/6 male mouse (age 8–10 weeks)
was sacrificed on day 0 (sham), day 2, and 10 after the surgery.
Kidneys were snap-frozen immediately after sacrifice. Pdgfrb-BAC-eGFP
reporter mice (for staining experiments, age 8–12 weeks, C57BL/6) were
developed by N. Heintz (The Rockefeller University) for the GENSAT
project. Genotyping of all mice was performed by PCR. Mice were housed
under specific pathogen-free conditions at the University Clinic
Aachen. Pdgfrb-BAC-eGFP were sacrificed on day 10 after the surgery.
All animal experiment protocols were approved by the LANUV-NRW,
Düsseldorf, Germany. All animal experiments were carried out in
accordance with their guidelines.
UUO scATAC-seq experiments
Nuclei isolation was performed as recommended by 10X Genomics
(demonstrated protocol [307]CG000169). The nuclei concentration was
verified using stained nuclei in a Neubauer chamber with trypan-blue
targeting a concentration of 10,000 nuclei. Tn5 incubation and library
prep followed the 10X scATAC protocol. After quality check using
Agilent BioAnalyzer, libraries were pooled and run on a NextSeq in
2 × 75 bps paired-end run using three runs of the NextSeq 500/550 High
Output Kit v2.5 Kit (Illumina). This results in more than 600 million
reads.
UUO data pre-processing
We used Cell-Ranger ATAC (v1.1.0) pipeline to perform low-level data
processing
([308]https://support.10xgenomics.com/single-cell-atac/software/pipelin
es/latest/algorithms/overview). We first demultiplexed raw base call
files using cellranger-atac mkfastq with its default setting to
generate FASTQ files for each flowcell. Next, cellranger-atac count was
applied to perform read trimming, filtering, and alignment. We then
estimated the transcription start site (TSS) enrichment score using the
obtained fragment files and filtered low-quality cells using a TSS
score of 8 and a number of unique fragments of 1000 as thresholds. The
obtained barcodes are considered valid cells for the following
analysis.
UUO data dimension reduction, data integration, and clustering
We next performed peak calling using MACS2 for each sample and merged
the peaks to generate a union peak set, which was used to create a peak
by cell matrix. For comparison, we applied distinct methods, i.e.,
scOpen, cisTopic, SnapATAC, and LSI/Cusanovich2018, to the matrix and
used the dimension reduced matrix for data integration, clustering, and
visualization. Next, we used Harmony^[309]31 to integrate the
scATAC-seq profiles from different conditions (day 0, day 2, and day
10) using either LSI/Cusanovich2018, cisTopic, scOpen, or SnapATAC
dimension reduced matrix as input. Specifically, we created a Seurat
object for each of the low-dimension matrices and ran the Harmony
algorithm with the function RunHarmony. We then used k-medoids to
cluster the cells taking batch-corrected low-dimension matrix as input.
The number of clusters was set to 17 given that the single-nucleus
RNA-seq that we used as a reference for annotation identified 17 unique
cell types (see below).
UUO label transfer
To evaluate and annotate the clusters obtained from data integration,
we downloaded a publicly available snRNA-seq dataset of the same
fibrosis model ([310]GSE119531) and performed label transfer using
Seurat3^[311]33. This dataset contains 6147 single-nucleus
transcriptomes with 17 unique cell types^[312]32. For label transfer,
we used the gene activity score matrix estimated by ArchR and
transferred the cell types from the snRNA-seq dataset to the integrated
scATAC-seq dataset by using the function FindTransferAnchors and
TransferData in Seurat3^[313]33. For benchmarking purposes, the
predicted labels were used as true labels to compute ARI for evaluation
of the clustering results and silhouette score for evaluation distances
after using different dimension reduction methods as input for data
integration (Supplementary Fig. [314]7c–e). We also performed the same
analysis for each sample separately and evaluated the results
(Fig. [315]3a).
UUO cluster annotation
For the biological interpretation, we estimated doublet scores using
ArchR^[316]34 and removed cells with a doublet score higher than 2.5.
Next, we named the cluster by assigning the label with the highest
proportion of cells to the cluster and checking marker genes
(Supplementary Fig. [317]9a). In total, we recovered 16 unique cell
types from the 17 labels, as two clusters (2 and 17) were annotated as
TAL cells. Specifically, we denoted clusters 6, 1, 3 as proximal tubule
(PT) S1, S2, and S3 cells. We annotated cluster 2 as thick ascending
limb (TAL), cluster 5 as distal convoluted tubule (DCT), cluster 7 as
collecting duct-principal cell (CD-PC), cluster 8 as an EC, cluster 9
as connecting tubule (CNT), cluster 10 as an intercalated cell (IC),
cluster 11 as fibroblast, cluster 12 as descending limb + thin
ascending limb (DL and TAL), cluster 13 as macrophage (MAC), cluster 16
as podocytes (Pod). Cluster 14 was identified as injured PT, which was
not described in ref. ^[318]32, given the increased accessibility of
marker Vcam1 and Havcr1 (Supplementary Fig. [319]9a). We also renamed
the cells of cluster 15, which were label as Mac2 in ref. ^[320]32, as
lymphoid cells given that these cells express B and T cell markers Ltb
and Cd1d, but not macrophage markers C1qa and C1qb. Finally, cluster 4
was removed based on the doublet analysis.
UUO Cell-type-specific footprinting with HINT-ATAC
We have adapted the footprinting-based differential TF activity
analysis from HINT-ATAC for scATAC-seq. In short, we created pseudo
bulk atac-seq libraries by combining reads of cells for each cell type
and performed footprinting with HINT-ATAC. Next, we predicted TF
binding sites by motif analysis (FDR = 0.0001) inside footprint
sequences using RGT (v0.12.3;
[321]https://github.com/CostaLab/reg-gen). Motifs were obtained from
JASPAR Version 2020^[322]73. We measured the average digestion profiles
around all binding sites of a given TF for each pseudo-bulk ATAC-seq
library. We used then the protection score^[323]4, which measures the
cell-specific activity of a factor by considering the number of
digestion events around the binding sites and depth of the footprint.
Higher protection scores indicate higher activity (binding) of that
factor. Finally, we only considered TFs with more than 1000 binding
sites and variance in activity score higher than 0.3. We also performed
smoothing for visualization of average footprint profiles. In short, we
performed a trimmed mean smoothing (5 bps window) and ignored cleavage
values in the top 97.5% quantile for each average profile.
Identifying trajectory from fibroblast to myofibroblast
We performed further sub-clustering of fibroblast cells on
batch-corrected low-dimension scOpen matrix. In total, 3 clusters were
obtained and annotated as pericyte (cluster 1), myofibroblast (cluster
2), and Scara5+ fibroblast (cluster 3) using known marker genes
(Supplementary Fig. [324]10a), respectively. For visualization, a
diffusion map 2D embedding was generated using R package
density^[325]74. Next, a trajectory from Scara5+ fibroblast to
myofibroblast was created using function addTrajectory and visualized
using function plotTrajectory from ArchR (Supplementary Fig. [326]10c).
Identifying key TF drivers of myofibroblast differentiation
To identify TFs that drive this process, we first performed peak
calling based on all fibroblasts using MACS2 to obtain specific peaks
and then estimated motif deviation per cell using chromVAR. The
deviation scores were normalized to allow for comparison between TFs.
Next, we selected the TFs with high variance of deviation and gene
activity score along the trajectory and calculated the correlation of
TF activity and gene activity. This was done by using the function
correlateTrajectories from ArchR. We only consider the 31 TFs with
significant correlation (FDR < 0.1) (Fig. [327]4c). We then sorted the
TFs by correlation, which identifies Runx1 as the most relevant TF for
the differentiation (Supplementary Fig. [328]10d).
Prediction of peak-to-gene links
We obtained TSS from annotation BSgenome.Mmusculus.UCSC.mm10 for each
gene and extended it by 250 kbps for both directions. Then, we
overlapped the peaks from fibroblasts and the TSS regions using
function findOverlaps to identify putative peak-to-gene links. We next
created 100 pseudo-bulk ATAC-seq profiles by assigning each cell to an
interval along the trajectory of myofibroblast differentiation. The
gene score matrix and peak matrix were aggregated according to the
assignment to generate two pseudo-bulk data matrices. For each putative
peak-to-gene link, we calculated the correlation between peak
accessibility and gene activity. The p-values are computed using t
distribution and corrected by Benjamini–Hochberg method. For
comparison, we also performed matrix imputation using the four top
methods, i.e., scOpen, SCALE, MAGIC, and cisTopic, as evaluated by
peaks recovering (Supplementary Fig. [329]2b) and computed the
correlation based on the imputed matrix.
To compare the scOpen predicted peak-to-gene correlation from different
types of peaks, we used the annotation generated by R package
ArchR^[330]34 and classified the peaks as distal, exonic, intronic, and
promoter. We also tested if the correlation is different between
activate enhancers and nonenhancers. For this, We obtained H3K27ac
([331]ENCSR000CDG) and H3K4me1 ([332]ENCSR436FYE) ChIP-seq peaks of
mouse kidneys from ENCODE. The peaks were classified as active
enhancers if they are overlapping with H3K27Ac and H3K4me1, and other
active regions if they are only overlapping with H3K27Ac.
Prediction and evaluation of Runx1 target genes
With each peak being associated with genes, we next sought to link
Runx1 to its target genes. For this, we first performed a footprinting
pseudo-bulk ATAC-seq profile to identify TF footprints inside peaks
linked to genes in the previous peak-to-gene analysis. Next, we
identified Runx1-binding sites using a motif-matching approach. We
defined the genes that have at least one footprint-support binding site
of Runx1 in their associated peaks as Runx1 target genes. We then used
the peak-to-gene correlation as a prediction between Runx1 and the
target genes. This procedure was for the peak to gene links predicted
by distinct imputation approaches, thus generating various predictions.
To evaluate the results, we used the DE genes obtained from RNA-seq of
Runx1 overexpression as true labels (see below), and computed the AUPR
(Fig. [333]4h).
Immunofluorescence staining of Runx1
Mouse kidney tissues were fixed in 4% formalin for 2 h at RT and frozen
in OCT after dehydration in 304%4 sucrose overnight. Using 5–10 μm
cryosections, slides were blocked in 5% donkey serum followed by 1-h
incubation of the primary antibody, washing three times for 5 min in
PBS, and subsequent incubation of the secondary antibodies for 45 min.
Following DAPI (4′,6-diamidino-2-phenylindole) staining (Roche,
1:10,000) the slides were mounted with ProLong Gold (Invitrogen,
#[334]P10144). Cells were fixed with 3% paraformaldehyde followed by
permeabilization with 0.3% TritonX. Cells were incubated with primary
antibodies and secondary antibodies diluted in 2% bovine serum albumin
in PBS for 60 or 30 min, respectively. The following antibodies were
used: anti-Runx1 (HPA004176, 1:100, Sigma-Aldrich), AF647 donkey
anti-rabbit (1:200, Jackson Immuno Research).
Confocal imaging and quantification
Images were acquired using a Nikon A1R confocal microscope using ×40
and ×60 objectives (Nikon). Raw imaging data were processed using Nikon
Software or ImageJ. Systematic random sampling was applied to the
sub-sample of at least three representative areas per image of
PDGFRbeGFP mice (n = 3 mice per condition). Using QuPath nuclei were
segmented and fluorescent intensity per nuclear size was measured of
PDGFRbeGFP positive nuclei.
Ethics
The ethics committee of the University Hospital RWTH Aachen approved
the human tissue protocol for cell isolation (EK-016/17). Kidney
tissues were collected from the Urology Department of the University
Hospital Eschweiler from patients undergoing nephrectomy due to renal
cell carcinoma.
Generation of a human PDGFRb+ cell line
The cell line was generated using MACS separation (Miltenyi biotec,
autoMACS Pro Separator, #130-092-545, autoMACS Columns #130-021-101) of
PDGFRb+ cells that were isolated from the healthy part of the kidney
cortex after nephrectomy as previously described in ref. ^[335]43. The
following antibodies were used for staining the cells and MACS
procedure: PDGFRb (R&D #MAB1263 antibody, dilution 1:100) and
anti-mouse IgG1-MicroBeads solution (Miltenyi, #130-047-102). The cells
were cultured in DMEM media (Thermo Fisher #31885) added 10% FCS and 1%
penicillin/streptomycin for 14 days. For immortalization (SV40-LT and
HTERT) the retroviral particles were produced by transient transfection
of HEK293T cells using TransIT-LT (Mirus). Amphotropic particles were
generated by co-transfection of plasmids pBABE-puro-SV40-LT (Addgene
#13970) or xlox-dNGFR-TERT (Addgene #69805) in combination with a
packaging plasmid pUMVC (Addgene #8449) and a pseudotyping plasmid
pMD2.G (Addgene #12259) respectively. Using Retro-X concentrator
(Clontech) 48 h post-transfection the particles were concentrated. For
transduction, the target cells were incubated with serial dilutions of
the retroviral supernatant (1:1 mix of concentrated particles
containing SV40-LT or rather hTERT) for 48 h. At 72 h after
transfection, the infected PDGFRb+ cells were selected with 2 μg/ml
puromycin for 7 days.
Retroviral overexpression of Runx1
Runx1 vector construction and generation of stable Runx1-overexpressing
cell lines. The human cDNA of Runx1 was PCR amplified from 293T cells
(ATCC, CRL-3216) using the primer sequences 5′-atgcgtatccccgtagatgcc-3′
and 5′-tcagtagggcctccacacgg-3′. Restriction sites and N-terminal
1xHA-Tag have been introduced into the PCR product using the primer
5′-cactcgaggccaccatgtacccatacgatgttccagattacgctcgtatccccgtagatgcc-3′
and 5′-acggaattctcagtagggcctccacac-3′. Subsequently, the PCR product
was digested with XhoI and EcoRI and cloned into pMIG (pMIG was a gift
from William Hahn (Addgene plasmid #9044;
[336]http://www.addgene.org/9044/; RRID:Addgene_9044). Retroviral
particles were produced by transient transfection in combination with
packaging plasmid pUMVC (pUMVC was a gift from Bob Weinberg (Addgene
plasmid #8449)) and pseudotyping plasmid pMD2.G (pMD2.G was a gift from
Didier Trono (Addgene plasmid #12259;
[337]http://www.addgene.org/12259/; RRID:Addgene_12259)) using
TransIT-LT (Mirus). Viral supernatants were collected 48–72 h after
transfection, clarified by centrifugation, supplemented with 10% FCS
and Polybrene (Sigma-Aldrich, final concentration of 8 μg/mL) and
0.45 μm filtered (Millipore; SLHP033RS). Cell transduction was
performed by incubating the PDGFβ cells with viral supernatants for
48 h. eGFP-expressing cells were single-cell sorted. A complete list of
primers used in this study is provided in Supplementary Table [338]2.
RNA isolation, RNA-Seq library preparation, and sequencing
RNA was extracted according to the manufacturer’s instructions using
the RNeasy Kit (QIAGEN). For RNA-seq Illumina TruSeq Stranded Total RNA
Library Preparation Kit was used using 1000 ng RNA as input. Sequencing
libraries were quantified using Tapestation (Agilent) and Quantus
(Promega). Equimolar pooling of the libraries was normalized to 1.8 pM,
denatured using 0.2 N NaOH, and neutralized with 200 nM Tris pH 7.0
prior to sequencing. Final sequencing was performed on a NextSeq500/550
platform (Illumina) according to the manufacturer’s protocols
(Illumina, CA, USA).
Analysis of RNA-seq data
Pipeline nf-core/rnaseq^[339]75 was used to analyze RNA-seq data.
Briefly, reads were aligned to the hg38 reference genome using
STAR^[340]76 and gene expression was quantified with Salmon^[341]77.
Deferentially expressed genes were identified using DESeq2^[342]78. We
used an adjusted p-value of 1e−05 and log2 fold change of 1 as
thresholds to select the significant DE genes, which were used as true
labels to evaluate the Runx1 target gene prediction. GO-enrichment
analysis was performed R package gprofiler2 and we showed results for
biological process and pathways from Human Phenotype Ontology
(Supplementary Fig. [343]11e). The volcano plot was generated by using
the R package EnhancedVolcano.
Calculation of population-doubling level (PDL)
For determining PDL, PDGFRb cells overexpressing Runx1 (or as control
having genomically integrated the empty vector sequence) were passaged
in six-well plates at a density of 1.5 × 10(4) cells/well. Every 96 h
(at sub-confluent state), cells were harvested and counted in a
hemocytometer before being re-seeded at initial density.
Statistical analysis and reproducibility
All reported p-values based on multi-comparison tests were corrected
using the Benjamini–Hochberg method. The number of samples for each
group was chosen on the basis of the expected levels of variation and
consistency. The depicted immunofluorescence micrographs are
representative. All studies were performed at least three times, and
all repeats were successful.
Reporting summary
Further information on research design is available in the [344]Nature
Research Reporting Summary linked to this article.
Supplementary information
[345]Supplementary Information^ (42.6MB, pdf)
[346]Peer Review File^ (1.9MB, pdf)
[347]41467_2021_26530_MOESM3_ESM.pdf^ (379.8KB, pdf)
Description of Additional Supplementary Files
[348]Supplementary Data 1^ (9.4KB, xlsx)
[349]Supplementary Data 2^ (90.5KB, xlsx)
[350]Reporting Summary^ (2.2MB, pdf)
Acknowledgements