Skip to main content

SampleQC: robust multivariate, multi-cell type, multi-sample quality control for single-cell data

Abstract

Quality control (QC) is a critical component of single-cell RNA-seq (scRNA-seq) processing pipelines. Current approaches to QC implicitly assume that datasets are comprised of one cell type, potentially resulting in biased exclusion of rare cell types. We introduce SampleQC, which robustly fits a Gaussian mixture model across multiple samples, improves sensitivity, and reduces bias compared to current approaches. We show via simulations that SampleQC is less susceptible to exclusion of rarer cell types. We also demonstrate SampleQC on a complex real dataset (867k cells over 172 samples). SampleQC is general, is implemented in R, and could be applied to other data types.

Background

Developments in single-cell RNA-seq (scRNAseq) technology have enabled experimenters to quantify transcriptional profiles for ever larger and more complex experiments [1,2,3]. The transformational nature of single-cell RNA-seq is its ability to measure transcriptional expression from nanolitre volumes. However, these tiny volumes result in noisy measurement, and the cells themselves can be strongly affected by the process of measurement. Quality control (QC) is therefore a critical element of pre-processing scRNAseq data: experimenters wish to carefully remove “bad” cells, while minimizing both loss of healthy cell datapoints and any bias in the types of cells retained [4]. Cells can be “bad” due to either technical issues such as droplets with unsuccessful reactions, or biological perturbations such as apoptosis. As the size and complexity of experiments increases, doing this for each sample individually becomes more challenging, and researchers require computational assistance to make the task feasible.

A fundamental issue with methods aiming to exclude poor quality cells is adequately defining “poor quality.” This is both challenging in itself and context-dependent. One approach to defining quality is in terms of the quantity of information provided by a cell, i.e., the number of reads or features. We might then seek to exclude all cells with too few reads. However, if there are sufficiently many of these cells, the total information provided could be enough to perform useful tasks (e.g., using “pseudobulk” approaches to identify genes with differential expression between conditions [5, 6]). Similarly, high proportions of mitochondrial reads are associated with stressed and apoptotic cells [7], and excluding cells on the basis of high mitochondrial proportion (e.g., cells with greater than 5% mitochondrial reads in mice, or greater than 10% in human cells; [8]) is a recommended approach to QC [4, 9]. However, analysis of healthy tissue shows that the proportion of mitochondrial reads shows strong variation across both species and tissue type [8], suggesting that this fixed-threshold strategy may exclude healthy cells and fail to exclude relatively stressed cells. Users may prefer more sensitive approaches that do not give hard assessments of cell quality, but give them the power to decide which are the unwanted cells.

Based on the various tutorials available, the current industry standards for QC are scater [10] and Seurat [11], both of which are data-driven rather than based on a fixed threshold. In addition, a probabilistic method miQC was recently introduced [12]. All three of these are unimodal (scater and Seurat are also univariate in typical use). scater and Seurat function by calculating robust measures of center and scale (such as median and median absolute deviation, or MAD) for each QC metric, then discarding cells that are a specified number of deviations away from the centers. They are typically applied individually to each sample, which we find may introduce bias. Most importantly, where samples are composed of multiple cell types whose QC metric distributions differ (e.g., they have lower counts, or higher mitochondrial proportions), such filtering will preferentially exclude cells from the cell types with more extreme values, leading to a bias towards excluding cells from particular cell types. This was noted by Germain et al., who found that stringent filtering on all cells simultaneously led to exclusion of cells that have extreme QC metrics, but not poor quality cells [13]. miQC is multivariate, assuming that the relationship between features detected and mitochondrial proportion is different in healthy cells and in “compromised” cells. This approach is also unimodal and may therefore also suffer from a bias against extreme cell types. Germain et al. also found that filtering was necessary, as lenient filtering led to poorer downstream performance (as measured by clustering). Taken together, these findings motivate the need for a QC filtering tool that allows for the presence of multiple cell types.

As the size of scRNAseq experiments increases, so does their complexity, resulting in ever-higher numbers of conditions and samples measured per experiment [14]. This increases the challenge of doing sensitive QC, e.g., by manually deciding a threshold for each sample. In addition, it is important to measure experimental quality at a sample and not just a cell level. An experimenter may wish to make comparisons between samples deriving from multiple experimental conditions; this requires establishing that there are sufficient high quality samples of each desired condition.

To address these problems, we have developed SampleQC, a computational method designed for QC of large scRNAseq experiments with complex designs. SampleQC is implemented as an R package based on a robust multivariate Gaussian mixture model (GMM). By fitting a mixture model in which each of the mixture components corresponds to one “QC cell type,” SampleQC identifies the QC distributions for multiple cell types simultaneously, removing the bias towards excluding healthy cell types with extreme QC metric values. We use a fitting procedure that is statistically robust, in the sense that it expects outliers to be present and inference is not distorted by them. By allowing each sample to have its own “sample shift” term, SampleQC fits simultaneously across multiple samples, which both increases statistical power and automates an otherwise tedious process. This addresses not only problems with sensitivity of outlier detection, but also provides sample-level QC outputs. Finally, SampleQC provides a comprehensive, automatically-generated report on cell and sample quality.

Results

SampleQC overview

Examining the distributions of QC metrics across many single-cell experiments, we observed that the values for a given sample would typically follow a multivariate Gaussian mixture distribution, i.e., groups of cells concentrated around distinct mean values, with distances from these mean values following approximately ellipsoid densities (Additional file 1: Fig. S1). We also observed that multiple samples often showed approximately the same distribution, subject to a sample-level shift in values, i.e., the individual values might be on average higher or lower than other samples, but the shapes of the ellipsoids and the relative distances between their means remained approximately the same (Additional file 1: Fig. S1). This motivated the following model to describe the metrics: for a cell i from sample j, which is a member of QC cell type k (k is not known), \(X_i \sim N( \alpha _0 + \alpha _j + \beta _k, \Sigma _{k})\), where \(X_i\) is the vector of QC metrics, \(\alpha _0\) is the global mean, \(\alpha _j\) is the mean shift of sample j relative to the global mean, and \((\beta _k, \Sigma _k)\) describe mean and variance-covariance of mixture component k. Finally, we observed that across large experiments, not all samples would follow the same mixture distribution, but that the samples could be grouped into “sample groups” with similar distributions of QC metrics.

For the metrics to follow a Gaussian mixture model, it is important that the marginal distributions of unimodal data are approximately Gaussian. This is dependent on appropriate transformations of the data, that allow non-Gaussian metrics to be transformed into spaces where they are approximately Gaussian. Applying log transformations to library sizes and genes detected is common practice; in addition, we have found that applying the logit (inverse logistic) transform to mitochondrial proportions often results in approximately Gaussian distributions (Additional file 1: Fig. S2). These observations suggest that, after appropriate transformations, a Gaussian mixture model may be a reasonable description of the data.

We assumed that the components of the mixture model represent “QC cell types,” where cells at the centers of the distributions are good quality, and cells that are unusually distant from these centers are cells with extreme QC metric values that should be discarded. In addition, we sought to identify entire samples that were very different from most samples, to flag these to the user for possible exclusion. This motivated an outlier detection algorithm based on the following two steps: (1) identifying groups of samples whose multivariate QC values approximately follow the same distributions; and (2) within each of these, fitting a multivariate GMM to identify cell-level outliers. These steps are outlined in Fig. 1.

Fig. 1
figure 1

SampleQC methodology. A Cartoon of cells and samples with varying quality, and their corresponding QC metrics. B SampleQC estimates distributional distances between samples and uses this to identify groups of samples. C SampleQC fits a robust GMM to each group of samples. D Low likelihood is used to exclude poor quality cells. E SampleQC generates report of outliers and overall sample quality

To identify groups of samples with similar distributions, SampleQC uses a non-parametric statistical measure of dissimilarity between multivariate samples, maximum mean discrepancy (MMD: [15]), to estimate “distances” between samples. MMD takes two samples as input, which must have the same dimensions but may have different numbers of observations, and calculates a measure of distance/dissimilarity that summarizes differences in means, (co)variances, and higher moments. We then use pairwise distances to construct a nearest neighbor graph that allows the identification of “sample groups,” i.e., groups of samples following approximately the same QC metric distributions. We use Louvain clustering to identify the groups [16]. The distances are also used as input to dimensionality reduction techniques, such as multidimensional scaling (MDS: [17]) and UMAP [18]. This gives low-dimensional embeddings where each point corresponds to a sample and permits users to check for batch effects or other issues with the data. For example, users can check the extent to which samples from the same batch are clustered together, potentially indicating sample quality problems with a batch.

The second step identifies cell outliers, i.e., cells with very different QC metric values to most other cells. Additionally, this step checks for outlier samples by estimating the mean shift in QC statistics for each sample. After SampleQC has identified sample groups, the user then determines how many components are present in each sample group by inspecting the QC distributions within each group, i.e., how many components the GMM should have. SampleQC then fits a robust GMM to the cells in each sample group.

Our GMM makes two main assumptions: (1) the configuration of QC cell types (i.e., their covariance matrices and relative means) is constant across the samples in a sample group; and (2) each sample has a uniform shift in (transformed) QC metrics. Our justification for these assumptions is the observation of distributions of QC metrics discussed above, and the empirical performance of the method, although reasonable biological arguments can also be made for them. The model allows the proportions of QC cell types (i.e., mixture components) to vary by sample; details of the mathematical model are discussed in the Methods section (see SampleQC method). This model structure, in which the same QC cell types are assumed to be present in every sample, has benefits when identifying outlier cells. The model learns the parameters for the QC cell-type components across the whole sample group, so that even when a sample has only a small proportion of a given QC cell type, these cells are not regarded as outliers. These cells may in fact be of particular interest, being from a cell type with reduced proportion relative to other samples.

We fit this model to identify poor quality cells, namely those that are outliers with respect to the distribution of the bulk of cells. While the presence of outliers is known to affect the estimation of statistics, especially higher order moments such as covariance [19], accurate estimation of the covariance matrices is essential precisely for identifying those outliers. We therefore use a robust estimation of the mean and covariance matrices [20]: robust estimators (like the MAD) give good but slightly less than optimal results when the data do follow an assumed distribution, but are also guaranteed to give good results when the data is contaminated and MLEs may fail [19]. We explicitly assume that outliers may be present, making this approach necessary.

Once we have fit models to each sample group, SampleQC is able to describe all cells in the included samples in terms of mixture components, which we assume to represent good quality cells. We can then identify outlier cells by calculating a statistical measure of distance from the center of each QC cell type (Mahalanobis distance, which indicates how unlikely a particular observation is under a \(\chi ^2\) distribution), and exclude cells that are unlikely to be a member of any component.

Simulating QC data for a large experiment

With single-cell data, there is no ground truth regarding which cells or samples are poor quality, making assessment of the performance of QC methods challenging. To evaluate SampleQC against other approaches, we therefore developed a simulation framework that replicates the distributions of QC metrics observed in biological datasets (pseudocode provided in Methods section; see the Simulations section). The framework and parameters are derived from large and complex experimental designs, including outliers, and are based on a hierarchical model.

At the experiment level, we make the following assumptions: (1) the experiment is composed of a number of sample groups with similar QC distributions; (2) there are a small number of “QC cell types” present in the experiment. By “QC cell type,” we mean a cluster of cells that is distinct within “QC space,” i.e., the selected QC metrics. We expect that multiple true cell types, i.e., clusters that are distinct in terms of expression, may have the same distribution in QC terms. These QC cell types are assumed to be common across all groups of samples, with a different subset present in each sample group. For each group, we randomly sample samples and cells, and which cell types are present. We also sample the means and covariances for the QC cell types, \(\{\beta _k, \Sigma _k\}\).

Within each sample group, we then model outliers and sample shifts at the level of samples. We assume that for each sample j, there is a uniform shift in the (transformed) QC metrics, \(\alpha _j\). This affects all cells in a sample equally. We also assume that each sample has different proportions of the QC cell types, sampled from a Dirichlet distribution. Each sample has a different proportion of outliers, modeled as a beta distribution, which gives all samples from the same group outlier proportions that vary around a common mean value. Outliers are assumed to differ from “good” cells by having lower non-mitochondrial reads and numbers of features, and correspondingly higher mitochondrial proportions; we model this by a sample-level parameter corresponding to the proportion of non-mitochondrial reads lost (modeled as a logistic normal).

For every cell, we have annotations of sample group (g), sample (j), and QC cell type (k), with corresponding values of global mean (\(\mu _g\)), sample shift (\(\alpha _j\)), and QC cell type mean and covariance (\(\beta _k\) and \(\Sigma _k\)). We draw a healthy QC metric vector for a given cell via a multivariate normal with mean \(\mu _g + \alpha _j + \beta _k\), and covariance \(\Sigma _k\). We then determine whether this cell is an outlier via a Bernoulli distribution, and if it is, we perturb this value by downsampling the non-mitochondrial reads and the QC metrics are updated accordingly.

This approach follows the model assumed by the GMM (SampleQC model), with two additional points to make the simulations more challenging. Firstly, the GMM does not include any outliers. To model these, we assume that each sample has a given proportion of outlier cells, selected at random. For each of these cells, the non-mitochondrial reads and the number of features are decreased by a given proportion, according to a beta distribution. This results in both a reduction in total counts and features, and an increase in mitochondrial proportion, consistent with outliers observed in real data. Secondly, we assume that the relative positions of the components, \(\{\beta _k\}\), are not identical in each sample, and vary by a small amount, \(\delta _{jk}\).

This results in simulated QC metrics for tens of thousands of cells across tens of samples, where the distributions of these metrics vary coherently across different groups of samples. The simulations provide two matrices of QC metrics: one that mimics the QC values we might see from experiments, and one for the QC values of the cells “before” they became low quality (such a matrix would never be observed in real life).

Benchmarking SampleQC against scater and miQC on simulated data

We generated simulated data for an experiment with 100,000 cells across multiple samples, resulting in 64 samples with a median of 1300 cells per sample, including outliers. We then ran SampleQC, scater, and miQC to compare performance: both in terms of excluding outliers, and retaining “good” cells. Inspecting the distributions of the outliers identified by the different methods, we observe that where multiple QC cell types are present, SampleQC identifies outliers correctly, while both scater and miQC may preferentially exclude cells from one QC cell type as outliers (Fig. 2). Where only one QC cell type is present, SampleQC, scater and miQC show similar results (Additional file 1: Fig. S3), as expected.

Fig. 2
figure 2

QC method comparisons on multimodal simulated data. A Biaxial plot of QC metrics for one simulated sample. Each point represents the QC metrics for one cell, with (known) simulated outliers in red and non-outliers in gray. B Multivariate density plot of data in A, annotated with models fit by SampleQC. The distinct clusters here are examples of what we term “QC cell types,” i.e., cells with similar distributions in their QC metrics, although their gene expression distributions may be distinct. C Mahalanobis distances to nearest cluster under the fit SampleQC model, with large values indicating low likelihood under multivariate Gaussian mixture model. D, E, F Outliers detected by SampleQC, scater, and miQC, respectively. These indicate the different assumptions on outliers made by the different methods: SampleQC calls droplets in low-density regions of the QC metric space as outliers; scater calls droplets with extreme values on at least one metric as outliers, in effect drawing a box around the central mode of the data; miQC identifies droplets with high mitochondrial proportion relative to the number of features observed, which may correspond to a good quality cell type with high mitochondrial proportion

When we assess performance in identifying good cells across the entire simulated dataset, we find that SampleQC has equal or better performance than both scater and miQC for both precision and recall (Fig. 3A, Additional file 1: Fig. S4). The relative performance on individual samples is affected by how many cell types are present in the sample. Where the data is multimodal, the multimodal model in SampleQC gives it equal or better performance than scater and miQC for both precision and recall (Additional file 1: Figs. S4 and S5). Where the data is unimodal (i.e., has one mixture component), scater and SampleQC show similar performance (Fig. 3B). In this case, performance of miQC is slightly worse, potentially because it assumes that a reasonable quantity of “compromised” cells must be present; when there are only few such cells, this may result in “good” cells reported as outliers.

Fig. 3
figure 3

Performance comparison of SampleQC , scater, and miQC on simulated data. Results from a simulated experiment of 100k cells with 3 QC cell types. A Precision-recall curve of identification of “good” cells. Solid lines show curve calculated over all cells. Transparent lines show curves for individual samples. Dots show default exclusion thresholds for each method (\(< 1\%\) Chi-squared likelihood under fitted model for SampleQC; \(> 2.5\) MADs for scater; \(> 0.75\) posterior probability of compromised cell for miQC). B Bias in number of cells reported by sample, samples split by whether one or multiple QC cell types present in the sample (i.e., whether samples is uni- or multimodal). y-axis is bias in reporting of good cells

Importantly, SampleQC also shows better performance than the compared methods in terms of cell-type bias (Additional file 1: Fig. S6). As above, when one QC cell type is present in a sample, SampleQC and scater have very similar biases in excluding good cells at default settings. However, when multiple QC cell types are present in a sample, SampleQC excludes similar numbers of good cells and shows low bias towards any particular QC cell type. In this setting, the good cells mistakenly excluded by both miQC and scater are almost entirely comprised of one cell type, while SampleQC’s more flexible model better preserves rare QC cell-type proportions. We note that these results are dependent on the choices made in developing the simulation and that alternative reasonable choices may give different results.

We performed the same comparisons for simulations where the data is t-distributed and found similar differences in performance between the methods (see dashed lines in Additional file 1: Fig. S4).

Number of QC clusters

Determining the appropriate number of distinct clusters in a dataset is a well-studied problem in statistics that remains challenging [21]. To avoid this problem, SampleQC requires users to specify the number of mixture components for each group of samples. We simulated experiments to test the sensitivity of our results to mismatches between the selected and true k. We simulated data with different true numbers of QC cell types, varying over 1, 2, and 3 QC cell types (our observation over many studies has been that many samples have between 1 and 4 mixture components). For each value of k_true, we simulated data for 20,000 cells divided into samples, with an average of 2000 cells per sample. We then fit SampleQC to each dataset, with the assumed number of clusters k_fit varying between 1 and 4. We found that a mismatch between k_true and k_fit could result in a reduction in performance (while SampleQC performed well when k_fit was correctly specified) (Additional file 1: Fig. S7). However, for multiple of the combinations where k_fit was misspecified, SampleQC was not able to fit; this provides some protection in itself. These results indicate that review of the diagnostic plots rendered by SampleQC is an important part of the QC process, allowing users to determine plausible values of k from the empirical distributions.

Applying SampleQC to complex biological datasets

We applied SampleQC to a large single-nuclei RNA-seq dataset comprising 867k cells over 172 samples (see Availability of data and materials), taken from human brain tissue, including samples from patients with neurodegenerative conditions. This illustrates an intended use case for SampleQC: a dataset with sufficiently many samples to make it challenging to adjust outlier thresholds for each sample individually, and where complex batch effects could be present. Here, we use log library size, logistic transform of mitochondrial proportion, and log2(spliced reads/unspliced reads) as QC metrics. The motivation for using log splice ratio in single nuclei data comes from the difference in abundance of unspliced reads between the nucleus and the cytoplasm [22]. In principle, unspliced reads should only be present in the nucleus; high ratios of spliced to unspliced read counts therefore allow us to identify nuclei that were not successfully stripped of their cytoplasm and are likely to be contaminated. In addition, this demonstrates the flexibility of SampleQC, which only assumes that multiple QC cell types are present, and does not require specific metrics to be used.

SampleQC first identifies samples with similar QC distributions, by calculating pairwise MMD distances between the samples. The principle reason for this is to allow SampleQC to fit separate models to each group of individual samples. Low dimensional projections of the resulting dissimilarities are shown in Fig. 4 (UMAP) and Additional file file 1: Fig. S8 (MDS), showing the groups identified by SampleQC (Fig. 4A), allowing investigators to assess whether sample source, sequencing run or other covariates might have influenced sample quality (Fig. 4B). Investigators can also check whether there are groups of samples that should be excluded entirely due to poor quality.

Fig. 4
figure 4

Sample-to-sample distance embedding (UMAP). UMAP embedding, based on sample-sample similarities calculated with MMD, resulting in samples with similar QC metrics being plotted close to each other. A Sample group, i.e., clusters of samples with similar QC metrics distributions, as estimated by SampleQC. Remaining plots show sample metadata and statistical summaries: B Sequencing batch, C mitochondrial proportion (categorized), D number of cells (categorized), E median counts per cell, F median log2 splice ratio per cell

Applying SampleQC to this dataset shows similar results to those for the simulation: where a cell population is present in the whole dataset but only makes up a small proportion of a sample, SampleQC is better able to preserve these cells than scater and miQC (Fig. 5). The Mahalanobis distances calculated under the SampleQC model also follow the expected distribution, i.e., most points follow a Chi-squared distribution with degrees of freedom equal to the number of dimensions, plus an outlier component with larger distances (Additional file 1: Fig. S9).

Fig. 5
figure 5

QC method comparisons on complex biological data. A Biaxial empirical density plot of QC metrics for single nuclei data, showing one selected sample, annotated with models fit by SampleQC. Color indicates density of cells with the same QC metrics. B Mahalanobis distances to nearest cluster under fit SampleQC model, with large values indicating low likelihood under multivariate Gaussian mixture model. C, D, and E Outliers detected by SampleQC, scater, and miQC, respectively. SampleQC outliers include user specification that QC cell types with mean splice ratio \(>3\) should be excluded

An alternative approach to QC for single-cell data is to apply only a minimal filter on QC metrics (to remove droplets that contain very little information and/or contain only lysed cells), cluster the cells as is normal for single cell, then remove clusters that show poor QC metrics. This approach works for some datasets, however it may become difficult to apply in more challenging samples. Applying this approach to single-cell data from ovarian cancer tissue [12], we found that while some of the clusters should clearly be excluded, other clusters showed a wide range of QC metrics (Additional file 1: Fig. S11). For example, clusters K06, K07, and K08 all include substantial numbers of cells with mitochondrial percentages both above 25% and below 10%, showing that this approach to excluding poor quality cells may be more difficult in samples not comprising healthy cells.

Although SampleQC is a more complex model than other approaches to QC, this does not substantially decrease speed. As an illustration, we timed SampleQC on the simulated dataset in Fig. 3, comprising 100k cells over 64 samples. The three steps of SampleQC took the following times: 44s to calculate pairwise MMDs; 70s to fit the SampleQC models; and 104s to render the report.

The methodology underlying SampleQC is general and not specific to scRNAseq. The example above demonstrates its application to single nuclei RNAseq, and in principle, it can identify outliers in any dataset comprising a small set of mixture components described by a small set of variables. This includes other single-cell technologies, such as CITE-seq, which is similar to scRNAseq but also quantifies protein expression via oligo-tagged antibodies [23], and scATACseq, which quantifies chromatin accessibility in single cells [24]. To demonstrate this, we ran SampleQC on CITE-seq data from PBMCs [25], using similar QC metrics to scRNAseq: log RNA counts; log RNA features; logistic transform of mitochondrial proportion; log ADT (protein) counts; log ADT features. The models fit by SampleQC successfully converged to the modes of the data and could be used for outlier detection (Additional file 1: Fig. S10).

Discussion

We introduced SampleQC, an approach for quality control of scRNAseq experiments that improves on the current industry standard and is designed for large and complex experimental designs. We show that our model simultaneously allows flexibility and sensitivity and reduces bias. Grouping samples by similarity of QC metric distributions allows a different model to be used for each sample group, which then increases sensitivity by borrowing strength across samples. However, SampleQC (as with other QC methods) is still not able to know whether a cell listed as an outlier is one because of technical or biological reasons.

One of the main challenges in this work is the lack of ground truth to evaluate methods. For this reason, previous QC filtering methods have not performed comprehensive benchmarks. Meanwhile, most datasets are annotated to cell types after filtering and therefore not annotated as good/bad for such an assessment. And while filtering is necessary at some level, it is also not always straightforward to assign a reason for a cell’s exclusion. For example, many of the excluded cells according to SampleQC are in low-density regions, but it could be they are not too extreme in each univariate QC metric, but reside in a low-density region in a multivariate sense. Furthermore, none of the tools available for QC filtering, including SampleQC, perform rigorous validation that low-quality cells are truly removed.

Our method is based on empirical observations of real data and is motivated by known biological mechanisms. Many single-cell techniques assume that scRNAseq data comprises multiple cell types that are distinct in terms of gene expression, however this is not reflected in current approaches to QC. Metrics used for quality control are functions of the full expression vector of counts. When multiple cell types are present in the full expression data, our observations over multiple datasets are that at least some separation into “QC cell types” is typically preserved in the restricted “QC space.” Our motivations regarding outliers, i.e., that they form a small proportion of the dataset with lower read counts and higher mitochondrial proportions, are also consistent with this observation. SampleQC allows users to choose any metric for input, and if the selected metrics approximately follow a GMM, the approach should produce sensible outputs. A GMM is a reasonable model given that single-cell data is known to often form clusters, and we have shown that the individual metrics (after appropriate transformations) typically follow Gaussian distributions. Non-Gaussian and non-parametric approaches to outlier detection could also be considered, such as estimating cell densities in QC space and then excluding cells in sparsely populated regions. However, this would risk excluding cells that were only rare in that sample, and our observations suggest that a GMM is reasonable for various experiments. By fitting one model across multiple samples, SampleQC is able to preserve cell types that are abundant in some samples, but have low abundance in others and are therefore discarded by methods that operate only at the sample level.

Although the downstream effects of choice of QC metrics are not well understood, there are good biological motivations for the metrics currently in use. Low library sizes indicate droplets where the cell lysed before the reaction took place, droplets that contain only debris, or where the reaction was not successful, while high library sizes may correspond to multiple cells in one droplet. Number of features observed is highly correlated with library size, and therefore, the same mechanisms for cell quality are relevant. Mitochondrial proportion may be high in cells that are stressed, or that lysed partially before measurement (resulting in a loss of cytoplasmic RNA and corresponding increased proportion of mitochondrial RNA); in single nuclei RNA-seq, the process of preparing nuclei should wash away mitochondria, meaning that mitochondrial reads may indicate contamination by ambient RNA. The relative proportion of spliced to unspliced reads indicates the relative abundances of nuclear and cytoplasmic RNA in a droplet [22]. Unspliced reads should only be present in the nucleus; high ratios of spliced to unspliced read counts in single nuclei data therefore allow us to identify nuclei that were not successfully stripped of their cytoplasm and are likely to be contaminated; meanwhile, in single-cell RNA-seq data, high proportions of unspliced (nuclear) RNA may indicate lysed cells. We expect that as new technologies are developed, new QC metrics will accompany them.

We have demonstrated SampleQC on standard measures of cell quality. However, the model is flexible and can be applied to any set of metrics specified by the user that approximately follow a Gaussian mixture distribution. We also note transformations that allow non-Gaussian metrics to be transformed into spaces where they are more likely to be Gaussian. For example, SampleQC transforms mitochondrial proportions via the logit function, and spliced:unspliced ratios via log. SampleQC is effectively a general outlier detection technique for GMMs, and this should allow SampleQC to be applied to datasets with other or multiple modalities. We have applied SampleQC to CITEseq (Additional file 1: Fig. S10), but in principle, the framework can be applied to data from scATACseq [24], scCATseq [26], single-cell techniques based on combined modalities, and spatial transcriptomics, by selecting appropriate measures (e.g., antibody tag depths for CITE-seq, sparseness for scATAC-seq, etc.). More exploration may be required to identify the set of QC measures that are sufficient to filter questionable cells, also because many existing measures are highly correlated, which could mean they are redundant.

QC does not need to be conducted entirely at the filtering stage. Another option is to carry forward more cells to a clustering/annotation step and then filter out subpopulations that are of debatable quality, assuming some biological knowledge can be used to assign this. And here, it is difficult to know if there is a clear “optimal” path. On the one hand, including more sparser cells may have adverse effects on similarity calculations, estimation of highly-variable genes, or on the low dimensional projections that clustering is based on; on the other hand, SampleQC summaries (e.g., P-values representing cells in low-density regions of their QC space) could be used afterwards as indicators for removal of individual clusters. The effect of including sparse cells on direct cell type annotation methods is not well studied. Furthermore, it is not clear at what point entire samples should be removed from downstream analysis; while SampleQC gives useful visualizations to characterize the QC of many samples, a judgment call is still needed.

It is worth noting that our performance results depend on the choices made in implementing the simulation, largely because we are not aware of other frameworks to simulate scRNA-seq QC summaries. Ultimately, our simulation highlights that inference from our model is possible, but does not directly show that cells are “bad.” While we tried to represent parameters and phenomena observed based on our experience in large scRNA-seq datasets, further development of the simulation framework would be desirable. We note that as the underlying models for both scater and miQC are unimodal, any circumstance where there are multiple distinct healthy cell populations may result in a performance advantage for SampleQC.

We do not provide any statistical guarantees that SampleQC converges to the optimal solution. However, SampleQC does provide automatically generated and comprehensive reports that show the fitted model over the empirical density. The models defined are conceptually simple, and the reports should therefore allow users to check both the validity of the fits and the appropriate number of clusters for each group.

We also do not attempt to solve the problem of selecting the appropriate value of k; this is a challenging and extensively-discussed problem in statistics [21]. As noted above, the automatic reports allow users to inspect whether the model has fit appropriately. In addition, using robust estimators makes the procedure less sensitive to misspecification of the number of components: robust methods expect some of the data to be outliers, and therefore do not attempt to “explain” all data; non-robust methods attempt to explain the whole dataset, and where the number of components is misspecified, the estimated means may lie between components.

Conclusions

As experiments increase in size and complexity, the task of removing unwanted cells becomes correspondingly more challenging, especially if users would like to preserve rarer populations. SampleQC seeks to solve this problem, by first identifying groups of samples with similar QC distributions, then fitting an empirically motivated statistical model to the QC metric data for each group. The model is simple, meaning that it is fast to fit and easy to check. This allows users to obtain an overview of data quality for their entire experiment and to accurately identify cells for exclusion without having to make decisions regarding every sample individually. Our intention is to provide a fast, flexible, and easy-to-use method that allows investigators to do quality control with minimal loss of their valuable data.

Methods

SampleQC model

SampleQC is based on standard approaches to identifying GMMs, with two important changes. Firstly, the model includes terms that are sample-specific: each sample has a sample shift term, i.e., a uniform change in values applying to all cells in a given sample. In addition, while we assume that each sample has the same mixture components (= QC cell types), the proportion of these components may vary within each sample. For cell i, which comes from sample j in QC cell type k, this gives us the following model for the QC values \(X_i\):

$$\begin{aligned}&X_i \sim N( \alpha _0 + \alpha _j + \beta _k, \Sigma _{k}) \quad \text {and} \end{aligned}$$
(1)
$$\begin{aligned}&P( Z_i=k | s_i=j) = p_{jk} \end{aligned}$$
(2)
$$\begin{aligned} \text {where}&\\ s_i = j,&\quad \text {cell}\ i\ \text {is from sample}\ j \\ Z_i = k,&\quad \text {cell}\ i\ \text {is in mixture component}\ k \\ \alpha _j&\quad \text {sample shift for sample}\ j \\ \beta _k&\quad \text {mean for mixture component}\ k \\ \Sigma _k&\quad \text {covariance matrix for mixture component}\ k \\ p_{jk}&\quad \text {mixing proportions in sample}\ j \end{aligned}$$

Secondly, the fit is robust, in the sense that it is less sensitive to outliers. The standard approach to fitting GMMs is via expectation-maximization, where the method alternates between fixing the means and covariance matrices then identifying the expected values for the latent cluster variables (“expectation”), and fixing the latent cluster variables then calculating the maximum likelihood estimators (MLEs) for the means and covariances (“maximization”). This approach assumes that all observations come from the GMM distribution. If the data is contaminated by a small proportion of observations that do not come from the assumed distribution (outliers), then this procedure may fail, especially regarding estimation of covariance matrices, in turn affecting cluster membership identification [19]. To mitigate this problem, we use a robust method to estimate the means and covariance matrices, based on using the points nearest the center of the component [20]. Robust estimators are designed to return good results even in the presence of outliers, which is especially important when the principle assumption of our task is that outliers may be present.

In addition to identifying outlier cells, fitting the GMM gives in estimated statistics as outputs, namely: the QC metric shift for each sample (\(\hat{\alpha }_j\)), the location and shape of each mixture component (\((\hat{\beta }_k, \hat{\Sigma }_k)\)), and the proportions of the components in each sample (\(p_{jk}\)). These can be used to identify samples with markedly different statistics to others in their sample group, both in terms of mean QC values and in terms of the split of cells across QC cell types. These samples are therefore flagged to experimenters for detailed checking and potential exclusion.

Criteria for outliers

In the software (see below), the user can also specify the threshold of the \(\chi ^2\) distribution to determine the how stringent the outlier exclusion should be. In particular, under the null of no outliers, the Mahalanobis distance (to the center of the nearest GMM component) should follow a \(\chi ^2\) distribution (with degrees of freedom equal to the number of QC metrics used as input). Large distances are therefore by definition very unlikely under the GMM model and represent the candidates to filter out. This can be achieved by setting a threshold on the tail of the \(\chi ^2\) distribution

Software

The input to SampleQC is a data.frame where each row corresponds to a cell; the columns must contain the chosen QC metrics and sample labels and can optionally also include additional sample annotations such as batch or experimental condition. Using a data.frame as input makes the software accessible to users of both the SingleCellExperiment and Seurat packages (and, by saving intermediate files as CSV, to users of scanpy). Please see the user guide for further details.

Prior to running SampleQC, we recommend excluding empty droplets, for example by running emptyDrops with either the default or a more generous cutoff, to ensure that all genuine cells are included. This may result in some empty droplets being used as input to SampleQC, but they should be identified as outlier cells and later excluded.

We then recommend removing multiplets before running SampleQC for two principal reasons. Firstly, excluding low quality cells first assumes that all doublets are composed of two good quality cells; by excluding low quality cells first, it is then more difficult to detect doublets comprising one good quality and one low quality cell. Secondly, just as the count vectors for doublets are linear combinations of two true cells, their QC metric values are also functions of combinations of two true cells. The practical impact of this is that doublets introduce a small amount of noise to the QC metric distributions; excluding them results in cleaner distributions that are slightly easier for SampleQC to fit. (In addition, the number of cells used in the run is needed to accurately estimate the expected number of doublets. Excluding low quality cells first would distort this calculation; although this can be addressed by recording the initial number of cells.)

To run SampleQC, the user first runs the function calculate_pairwise_mmds, which estimates MMD values for each pair of samples, identifies groups of samples with similar QC metric distributions, and calculates embeddings in lower-dimensional spaces (both MDS and UMAP). The user can then plot the samples and check for evidence of batch effects, e.g., if all samples from a given well, run or experimental condition are grouped together. Users can also check whether any sample group has consistently low quality cells (e.g., high mitochondrial proportions) and consider excluding all samples in that group.

To run the cell outlier section of SampleQC, the user then selects qc_names, the QC metrics to use, and k, a vector corresponding to the number of mixture components for each sample group, and calls the function fit_sampleqc. SampleQC then fits a GMM to each sample group, and uses cell likelihoods to identify outlier cells. Users would typically start by using \(k=1\) for all sample groups; as the simplest model, this is extremely fast. The report includes biaxial plots of the QC metrics for all samples, which allow users to judge what value of k is most appropriate for each sample group, then rerun SampleQC. Users can inspect the cells identified as outliers by generating an html report with the function make_sampleqc_report. The user can also specify alpha, the \(\chi ^2\) distribution threshold determining the how stringent outlier exclusion should be; however, this is less critical and the default value (alpha = 0.01) is generally appropriate. alpha specifies the distance away from the center of the nearest GMM component that should be considered as an outlier. Under a robust model, the distribution is assumed to be a GMM plus a contamination component that does not follow the GMM and is therefore by definition very unlikely under the GMM distribution, so as long as alpha is set reasonably low (e.g., 1%, 0.1%), this will only have marginal effects on results.

If a large proportion of a sample is of poor quality, SampleQC may identify these cells as one sample cluster, and one of the mixture model components may therefore be centered on them (e.g., the cells with high splice ratio in Fig. 5). Without intervention, these would be reported as good cells rather than outliers. SampleQC therefore includes functionality allowing users to specify that any component whose mean meets a certain criteria should be regarded as outliers. For example, a component whose mean mitochondrial percentage is greater than a given percentage can be labeled as outliers.

Simulations

We developed a framework for simulating samples of QC data that reflects the complexity observed in real datasets, following the model given in Eq. (1), but additionally modeling outliers; in the SampleQC R package, users can access this functionality from the simulate_qcs function. For the simulations, we assume that there are three QC metrics: log library sizes, log feature counts, and logit mitochondrial proportion. The simulations are hierarchical: firstly, parameters for group size and GMM components are drawn at the whole-experiment level; then, we draw parameters for sample quality at the sample group level, capturing variability that is coherent across groups of samples; then we draw parameters for each individual sample and QC metrics for each individual cell. To model the QC metrics, we assume that most cells are healthy, i.e., they derive from the experiment-level GMM, while a small number are “bad,” i.e., their values are perturbed away from the means of the GMM. These steps are set out in Algorithm 1.

figure a

Algorithm 1 simulate_qcs: Overview of simulations (* indicates variables that would be known and used as input for real data)

At the experiment level, we simulate the covariance matrices and relative means of the mixture components (QC cell types) that are present in the experiment (Algorithm 2). We then simulate parameters reflecting coherent behavior across different groups of samples. First, we partition the cells between the groups according to a multinomial distribution, simulate how many samples are present in each group, and randomly assign QC cell types to each group. We assign the QC cell types such that each group has at least one QC cell type present, and that no two groups have identical sets of QC cell types. To simulate coherent differences in outliers between each group, we simulate parameters from a beta distribution that determines how the proportion of outliers varies across samples (p_out_0, theta_0) and how perturbed these outliers will be relative to healthy cells (p_loss_0, the mean proportion of non-mitochondrial reads that is lost).

figure b

Algorithm 2 draw_expt_level: Pseudocode for drawing experiment-level parameters

We then simulate samples and cells within each sample group (Algorithm 4). Within each sample, we first simulate the sample shift from a multivariate Gaussian and simulate the proportions of the different QC cell types via a Dirichlet distribution (p_jk). We also simulate what proportion of cells in this sample are outliers, p_out_j, from a beta distribution with parameters (p_loss_0, theta_0), and the extent of perturbation in unhealthy cells, p_loss_j, from a logit-normal distribution.

We can then simulate each healthy cell, by first drawing its QC cell type from a multinomial with \({\textbf {p}} = \{\texttt {p\_jk}\}\), then drawing from multivariate normal \((\texttt {beta\_k}, \texttt {Sigma\_k})\) for the appropriate k. Adding this to the relevant mu_0 gives us the QC metrics for a healthy cell. We then decide whether this cell is healthy or not by drawing from a binomial distribution (with probability \(\texttt {p\_out\_j}\). If the cell is healthy, its QC metrics are left unchanged. If not, its non-mitochondrial reads are randomly decreased by a proportion p_loss_j. This results in both a reduction in total counts and an increase in mitochondrial proportion. We apply the same random reduction to the number of features.

figure c

Algorithm 3 draw_group_level: Pseudocode for drawing sample group-level parameters

figure d

Algorithm 4 draw_cell_level: Pseudocode for drawing cell-level parameters

The outputs from the simulations show similar distributions of QC metrics to real data (Fig. 5).

Datasets

The datasets listed in Table 1 were used in this paper, obtained via the scRNAseq package [27].

Table 1 List of datasets used in paper. All datasets are single-cell RNA-seq unless otherwise stated

Availability of data and materials

The data in Figs. 4 and 5 is QC data taken from human brain tissue, including samples from patients with neurodegenerative conditions. The data derives from a study undertaken as part of a research collaboration agreement between the University of Zürich and F. Hoffman La Roche AG, now published as a preprint [33]. Although we have made the anonymized QC metric data available [34], the full expression data is not yet public.

Code availability

The SampleQC software package is accessible at https://github.com/wmacnair/SampleQC; a code snapshot of the software package is available on zenodo [35]. A repo for reproducing the analyses in the paper is accessible at https://github.com/wmacnair/SampleQC_paper_analyses and the corresponding HTML reports are available from https://htmlpreview.github.io/?https://github.com/wmacnair/SampleQC_paper_analyses/blob/main/docs/index.html; a code snapshot of the analysis repository at time of submission is available on zenodo [34].

References

  1. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009;6(5):377–82.

    Article  CAS  PubMed  Google Scholar 

  2. Macosko EZ, Basu A, Satija R, Nemesh J, Shekhar K, Goldman M, et al. Highly Parallel Genome-wide Expression Profiling of Individual Cells Using Nanoliter Droplets. Cell. 2015;161(5):1202–14.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Klein AM, Mazutis L, Akartuna I, Tallapragada N, Veres A, Li V, et al. Droplet Barcoding for Single-Cell Transcriptomics Applied to Embryonic Stem Cells. Cell. 2015;161(5):1187–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol. 2019;15(6):e8746. https://www.embopress.org/doi/full/10.15252/msb.20188746.

  5. Crowell HL, Soneson C, Germain P-L, Calini D, Collin L, Raposo C, et al. muscat detects subpopulation-specific state transitions from multi-sample multi-condition single-cell transcriptomics data. Nat Commun. 2020;11:6077.

  6. Squair JW, Gautier M, Kathe C, Anderson MA, James ND, Hutson TH, et al. Confronting false discoveries in single-cell differential expression. Nat Commun. 2021;12:5692.

  7. Márquez-Jurado S, Díaz-Colunga J, das Neves RP, Martinez-Lorente A, Almazán F, Guantes R, et al. Mitochondrial levels determine variability in cell death by modulating apoptotic gene expression. Nat Commun. 2018;9(1):389.

    Article  PubMed  PubMed Central  Google Scholar 

  8. Osorio D, Cai JJ. Systematic determination of the mitochondrial proportion in human and mice tissues for single-cell RNA-sequencing data quality control. Bioinformatics. 2021;37:963–7.

  9. Amezquita RA, Lun ATL, Becht E, Carey VJ, Carpp LN, Geistlinger L, et al. Orchestrating single-cell analysis with Bioconductor. Nat Methods. 2020;17(2):137–45.

    Article  CAS  PubMed  Google Scholar 

  10. McCarthy DJ, Campbell KR, Lun ATL, Wills QF. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics. 2017;33(8):1179–86.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Stuart T, Butler A, Hoffman P, Hafemeister C, Papalexi E, Mauck WM 3rd, et al. Comprehensive Integration of Single-Cell Data. Cell. 2019;177(7):1888-1902.e21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Hippen AA, Falco MM, Weber LM, Erkan EP, Zhang K, Doherty JA, et al. miQC: An adaptive probabilistic framework for quality control of single-cell RNA-sequencing data. PLoS Comput Biol. 2021;17:e1009290.

  13. Germain PL, Sonrel A, Robinson MD. pipeComp, a general framework for the evaluation of computational pipelines, reveals performant single cell RNA-seq preprocessing tools. Genome Biol. 2020;21(1):227.

    Article  PubMed  PubMed Central  Google Scholar 

  14. Svensson V, da Veiga Beltrame E, Pachter L. A curated database reveals trends in single-cell transcriptomics. Database [Internet]. 2020;2020. Available from: http://dx.doi.org/10.1093/database/baaa073

  15. Gretton A, Borgwardt KM, Rasch MJ, Schölkopf B, Smola A. A Kernel Two-Sample Test. J Mach Learn Res. 2012;13(Mar):723–73.

    Google Scholar 

  16. Blondel VD, Guillaume J-L, Lambiotte R, Lefebvre E. Fast unfolding of communities in large networks. J Stat Mech. IOP Publishing; 2008;2008:P10008.

  17. Torgerson WS. Multidimensional scaling: I. theory and method. Psychometrika. 1952;17(4):401–19.

    Article  Google Scholar 

  18. McInnes L, Healy J, Saul N, Großberger L. UMAP: Uniform Manifold Approximation and Projection. J Open Source Softw. The Open J. 2018;3:861.

  19. Huber PJ. Robust Statistics. Wiley; 2004. https://onlinelibrary.wiley.com/doi/book/10.1002/9780470434697.

  20. Rousseeuw PJ, Van Driessen K. A Fast Algorithm for the Minimum Covariance Determinant Estimator. Technometrics. 1999;41(3):212–23.

    Article  Google Scholar 

  21. McLachlan GJ, Lee SX, Rathnayake SI. Finite Mixture Models. Annu Rev Stat Appl. Annual Reviews. 2019;6:355–78.

  22. Alvarez M, Rahmani E, Jew B, Garske KM, Miao Z, Benhammou JN, et al. Enhancing droplet-based single-nucleus RNA-seq resolution using the semi-supervised machine learning classifier DIEM. Sci Rep. 2020;10(1):11019.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Stoeckius M, Hafemeister C, Stephenson W, Houck-Loomis B, Chattopadhyay PK, Swerdlow H, et al. Simultaneous epitope and transcriptome measurement in single cells. Nat Methods. 2017;14(9):865–8.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP, et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature. 2015;523(7561):486–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kotliarov Y, Sparks R, Martins AJ, Mulè MP, Lu Y, Goswami M, et al. Broad immune activation underlies shared set point signatures for vaccine responsiveness in healthy individuals and disease activity in patients with lupus. Nat Med. 2020;26(4):618–29.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Liu L, Liu C, Quintero A, Wu L, Yuan Y, Wang M, et al. Deconvolution of single-cell multi-omics layers reveals regulatory heterogeneity. Nat Commun. 2019;10(1):470.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Risso D, Cole M. scRNAseq: Collection of Public Single-Cell RNA-Seq Datasets. 2020. R package version 2.8.0. https://bioconductor.org/packages/release/data/experiment/html/scRNAseq.html.

  28. Campbell JN, Macosko EZ, Fenselau H, Pers TH, Lyubetskaya A, Tenen D, et al. A molecular census of arcuate hypothalamus and median eminence cell types. Nat Neurosci. 2017;20(3):484–96.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Weber LM, Hippen AA, Hickey PF, Berrett KC, Gertz J, Doherty JA, et al. Genetic demultiplexing of pooled single-cell RNA-sequencing samples in cancer facilitates effective experimental design. Gigascience [Internet]. 2021;10. Available from: http://dx.doi.org/10.1093/gigascience/giab062

  30. Shekhar K, Lapan SW, Whitney IE, Tran NM, Macosko EZ, Kowalczyk M, et al. Comprehensive Classification of Retinal Bipolar Neurons by Single-Cell Transcriptomics. Cell. 2016;166(5):1308-1323.e30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wang W, Vilella F, Alama P, Moreno I, Mignardi M, Isakova A, et al. Single-cell transcriptomic atlas of the human endometrium during the menstrual cycle. Nat Med. 2020;26(10):1644–53.

    Article  CAS  PubMed  Google Scholar 

  32. Zeisel A, Muñoz-Manchado AB, Codeluppi S, Lönnerberg P, La Manno G, Juréus A, et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science. 2015;347(6226):1138–42.

    Article  CAS  PubMed  Google Scholar 

  33. Macnair W, Calini D, Agirre E, Bryois J, Jäkel S, Kukanja P, et al. Single nuclei RNAseq stratifies multiple sclerosis patients into three distinct white matter glia responses [Internet]. bioRxiv. 2022 [cited 2022 Jun 24]. p. 2022.04.06.487263. Available from: https://www.biorxiv.org/content/10.1101/2022.04.06.487263v1

  34. Macnair W, Robinson M. wmacnair/SampleQC_paper_analyses: v0.1.0. Zenodo. 2022. https://doi.org/10.5281/zenodo.6414319.

  35. Macnair W, Robinson M. wmacnair/SampleQC: v0.6.6. Zenodo. 2022. https://doi.org/10.5281/zenodo.6414311.

Download references

Acknowledgements

We thank Helena Crowell, Pierre-Luc Germain, and Simone Tiberi for their valuable feedback on the manuscript and software. We thank Dheeraj Malhotra and Roche for the large experimental dataset (see “Availability of data and materials” below).

Peer review information

Andrew Cosgrove was the primary editor of this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

Review history

The review history is available as Additional files 2, 3, and 4.

Funding

This work was supported by the Swiss National Science Foundation (grant numbers 310030_175841, CRSII5_177208). MDR acknowledges support from the University Research Priority Program Evolution in Action at the University of Zurich. The funder played no role in the design of this study or in its execution.

Author information

Authors and Affiliations

Authors

Contributions

We use the CRediT taxonomy to define author contributions: WM: conceptualization, methodology, software, formal analysis, investigation, visualization, data curation, writing - original draft, writing - review and editing. MDR: supervision, methodology, writing - review and editing, funding acquisition. The authors read and approved the final manuscript.

Corresponding author

Correspondence to Will Macnair.

Ethics declarations

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Additional information

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary Information

Additional file 1.

Supplementary Figures.

Additional file 2.

Reviewer reports.

Additional file 3.

Authors’ response to first round of review.

Additional file 4.

Authors’ response to second round of review

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated in a credit line to the data.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Macnair, W., Robinson, M. SampleQC: robust multivariate, multi-cell type, multi-sample quality control for single-cell data. Genome Biol 24, 23 (2023). https://doi.org/10.1186/s13059-023-02859-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1186/s13059-023-02859-3

Keywords