scholarly journals miQC: An adaptive probabilistic framework for quality control of single-cell RNA-sequencing data

2021 ◽  
Author(s):  
Ariel A. Hippen ◽  
Matias M. Falco ◽  
Lukas M. Weber ◽  
Erdogan Pekcan Erkan ◽  
Kaiyang Zhang ◽  
...  

AbstractMotivationSingle-cell RNA-sequencing (scRNA-seq) has made it possible to profile gene expression in tissues at high resolution. An important preprocessing step prior to performing downstream analyses is to identify and remove cells with poor or degraded sample quality using quality control (QC) metrics. Two widely used QC metrics to identify a ‘low-quality’ cell are (i) if the cell includes a high proportion of reads that map to mitochondrial DNA (mtDNA) encoded genes and (ii) if a small number of genes are detected. Current best practices use these QC metrics independently with either arbitrary, uniform thresholds (e.g. 5%) or biological context-dependent (e.g. species) thresholds, and fail to jointly model these metrics in a data-driven manner. Current practices are often overly stringent and especially untenable on lower-quality tissues, such as archived tumor tissues.ResultsWe propose a data-driven QC metric (miQC) that jointly models both the proportion of reads mapping to mtDNA genes and the number of detected genes with mixture models in a probabilistic framework to predict the low-quality cells in a given dataset. We demonstrate how our QC metric easily adapts to different types of single-cell datasets to remove low-quality cells while preserving high-quality cells that can be used for downstream analyses.AvailabilitySoftware available at https://github.com/greenelab/miQC. The code used to download datasets, perform the analyses, and reproduce the figures is available at https://github.com/greenelab/mito-filtering.ContactStephanie C. Hicks ([email protected]) and Anna Vähärautio ([email protected])

2021 ◽  
Vol 17 (8) ◽  
pp. e1009290
Author(s):  
Ariel A. Hippen ◽  
Matias M. Falco ◽  
Lukas M. Weber ◽  
Erdogan Pekcan Erkan ◽  
Kaiyang Zhang ◽  
...  

Single-cell RNA-sequencing (scRNA-seq) has made it possible to profile gene expression in tissues at high resolution. An important preprocessing step prior to performing downstream analyses is to identify and remove cells with poor or degraded sample quality using quality control (QC) metrics. Two widely used QC metrics to identify a ‘low-quality’ cell are (i) if the cell includes a high proportion of reads that map to mitochondrial DNA (mtDNA) encoded genes and (ii) if a small number of genes are detected. Current best practices use these QC metrics independently with either arbitrary, uniform thresholds (e.g. 5%) or biological context-dependent (e.g. species) thresholds, and fail to jointly model these metrics in a data-driven manner. Current practices are often overly stringent and especially untenable on certain types of tissues, such as archived tumor tissues, or tissues associated with mitochondrial function, such as kidney tissue [1]. We propose a data-driven QC metric (miQC) that jointly models both the proportion of reads mapping to mtDNA genes and the number of detected genes with mixture models in a probabilistic framework to predict the low-quality cells in a given dataset. We demonstrate how our QC metric easily adapts to different types of single-cell datasets to remove low-quality cells while preserving high-quality cells that can be used for downstream analyses. Our software package is available at https://bioconductor.org/packages/miQC.


2021 ◽  
Author(s):  
Helena L Crowell ◽  
Sarah X Morillo Leonardo ◽  
Charlotte Soneson ◽  
Mark D Robinson

With the emergence of hundreds of single-cell RNA-sequencing (scRNA-seq) datasets, the number of computational tools to analyse aspects of the generated data has grown rapidly. As a result, there is a recurring need to demonstrate whether newly developed methods are truly performant - on their own as well as in comparison to existing tools. Benchmark studies aim to consolidate the space of available methods for a given task, and often use simulated data that provide a ground truth for evaluations. Thus, demanding a high quality standard for synthetically generated data is critical to make simulation study results credible and transferable to real data. Here, we evaluated methods for synthetic scRNA-seq data generation in their ability to mimic experimental data. Besides comparing gene- and cell-level quality control summaries in both one- and two-dimensional settings, we further quantified these at the batch- and cluster-level. Secondly, we investigate the effect of simulators on clustering and batch correction method comparisons, and, thirdly, which and to what extent quality control summaries can capture reference-simulation similarity. Our results suggest that most simulators are unable to accommodate complex designs without introducing artificial effects; they yield over-optimistic performance of integration, and potentially unreliable ranking of clustering methods; and, it is generally unknown which summaries are important to ensure effective simulation-based method comparisons.


2019 ◽  
Author(s):  
Haruka Ozaki ◽  
Tetsutaro Hayashi ◽  
Mana Umeda ◽  
Itoshi Nikaido

AbstractBackgroundRead coverage of RNA sequencing data reflects gene expression and RNA processing events. Single-cell RNA sequencing (scRNA-seq) methods, particularly “full-length” ones, provide read coverage of many individual cells and have the potential to reveal cellular heterogeneity in RNA transcription and processing. However, visualization tools suited to highlighting cell-to-cell heterogeneity in read coverage are still lacking.ResultsHere, we have developed Millefy, a tool for visualizing read coverage of scRNA-seq data in genomic contexts. Millefy is designed to show read coverage of all individual cells at once in genomic contexts and to highlight cell-to-cell heterogeneity in read coverage. By visualizing read coverage of all cells as a heat map and dynamically reordering cells based on diffusion maps, Millefy facilitates discovery of “local” region-specific, cell-to-cell heterogeneity in read coverage, including variability of transcribed regions.ConclusionsMillefy simplifies the examination of cellular heterogeneity in RNA transcription and processing events using scRNA-seq data. Millefy is available as an R package (https://github.com/yuifu/millefy) and a Docker image to help use Millefy on the Jupyter notebook (https://hub.docker.com/r/yuifu/datascience-notebook-millefy).


2020 ◽  
Author(s):  
Rui Hong ◽  
Yusuke Koga ◽  
Shruthi Bandyadka ◽  
Anastasia Leshchyk ◽  
Zhe Wang ◽  
...  

AbstractPerforming comprehensive quality control is necessary to remove technical or biological artifacts in single-cell RNA sequencing (scRNA-seq) data. Artifacts in the scRNA-seq data, such as doublets or ambient RNA, can also hinder downstream clustering and marker selection and need to be assessed. While several algorithms have been developed to perform various quality control tasks, they are only available in different packages across various programming environments. No standardized workflow has been developed to streamline the generation and reporting of all quality control metrics from these tools. We have built an easy-to-use pipeline, named SCTK-QC, in the singleCellTK package that generates a comprehensive set of quality control metrics from a plethora of packages for quality control. We are able to import data from several preprocessing tools including CellRanger, STARSolo, BUSTools, dropEST, Optimus, and SEQC. Standard quality control metrics for each cell are calculated including the total number of UMIs, total number of genes detected, and the percentage of counts mapping to predefined gene sets such as mitochondrial genes. Doublet detection algorithms employed include scrublet, scds, doubletCells, and doubletFinder. DecontX is used to identify contamination in each individual cell. To make the data accessible in downstream analysis workflows, the results can be exported to common data structures in R and Python or to text files for use in any generic workflow. Overall, this pipeline will streamline and standardize quality control analyses for single cell RNA-seq data across different platforms.


GigaScience ◽  
2019 ◽  
Vol 8 (12) ◽  
Author(s):  
Graham J Etherington ◽  
Nicola Soranzo ◽  
Suhaib Mohammed ◽  
Wilfried Haerty ◽  
Robert P Davey ◽  
...  

Abstract Background It is not a trivial step to move from single-cell RNA-sequencing (scRNA-seq) data production to data analysis. There is a lack of intuitive training materials and easy-to-use analysis tools, and researchers can find it difficult to master the basics of scRNA-seq quality control and the later analysis. Results We have developed a range of practical scripts, together with their corresponding Galaxy wrappers, that make scRNA-seq training and quality control accessible to researchers previously daunted by the prospect of scRNA-seq analysis. We implement a “visualize-filter-visualize” paradigm through simple command line tools that use the Loom format to exchange data between the tools. The point-and-click nature of Galaxy makes it easy to assess, visualize, and filter scRNA-seq data from short-read sequencing data. Conclusion We have developed a suite of scRNA-seq tools that can be used for both training and more in-depth analyses.


Author(s):  
Yue Zhang ◽  
Shunfu Mao ◽  
Sumit Mukherjee ◽  
Sreeram Kannan ◽  
Georg Seelig

AbstractAnalysis of single cell RNA sequencing (scRNA-Seq) datasets is a complex and time-consuming process, requiring both biological knowledge and technical skill. In order to simplify and systematize this process, we introduce UNCURL-App, an online GUI-based interactive scRNA-Seq analysis tool. UNCURL-App introduces two key innovations: First, prior knowledge in the form of cell type, anatomy, and Gene Ontology databases is integrated directly with the rest of the analysis process, allowing users to automatically map cell clusters to known cell types based on gene expression. Second, tools for interactive re-analysis allow the user to iteratively create, merge, or delete clusters in order to arrive at an optimal mapping between clusters and cell types.AvailabilityThe website is at https://uncurl.cs.washington.edu/. Source code is available at https://github.com/yjzhang/uncurl_app


2021 ◽  
Vol 26 (5) ◽  
pp. 772-789
Author(s):  
Yu Tian ◽  
Ruiqing Zheng ◽  
Zhenlan Liang ◽  
Suning Li ◽  
Fang-Xiang Wu ◽  
...  

2019 ◽  
Author(s):  
Tamim Abdelaal ◽  
Lieke Michielsen ◽  
Davy Cats ◽  
Dylan Hoogduin ◽  
Hailiang Mei ◽  
...  

AbstractBackgroundSingle cell transcriptomics are rapidly advancing our understanding of the cellular composition of complex tissues and organisms. A major limitation in most analysis pipelines is the reliance on manual annotations to determine cell identities, which are time-consuming and irreproducible. The exponential growth in the number of cells and samples has prompted the adaptation and development of supervised classification methods for automatic cell identification.ResultsHere, we benchmarked 20 classification methods that automatically assign cell identities including single cell-specific and general-purpose classifiers. The methods were evaluated using eight publicly available single cell RNA-sequencing datasets of different sizes, technologies, species, and complexity. The performance of the methods was evaluated based on their accuracy, percentage of unclassified cells, and computation time. We further evaluated their sensitivity to the input features, their performance across different annotation levels and datasets. We found that most classifiers performed well on a variety of datasets with decreased accuracy for complex datasets with overlapping classes or deep annotations. The general-purpose SVM classifier has overall the best performance across the different experiments.ConclusionsWe present a comprehensive evaluation of automatic cell identification methods for single cell RNA-sequencing data. All the code used for the evaluation is available on GitHub (https://github.com/tabdelaal/scRNAseq_Benchmark). Additionally, we provide a Snakemake workflow to facilitate the benchmarking and to support extension of new methods and new datasets (https://github.com/tabdelaal/scRNAseq_Benchmark/tree/snakemake_and_docker).


2020 ◽  
Vol 22 (Supplement_2) ◽  
pp. ii110-ii110
Author(s):  
Christina Jackson ◽  
Christopher Cherry ◽  
Sadhana Bom ◽  
Hao Zhang ◽  
John Choi ◽  
...  

Abstract BACKGROUND Glioma associated myeloid cells (GAMs) can be induced to adopt an immunosuppressive phenotype that can lead to inhibition of anti-tumor responses in glioblastoma (GBM). Understanding the composition and phenotypes of GAMs is essential to modulating the myeloid compartment as a therapeutic adjunct to improve anti-tumor immune response. METHODS We performed single-cell RNA-sequencing (sc-RNAseq) of 435,400 myeloid and tumor cells to identify transcriptomic and phenotypic differences in GAMs across glioma grades. We further correlated the heterogeneity of the GAM landscape with tumor cell transcriptomics to investigate interactions between GAMs and tumor cells. RESULTS sc-RNAseq revealed a diverse landscape of myeloid-lineage cells in gliomas with an increase in preponderance of bone marrow derived myeloid cells (BMDMs) with increasing tumor grade. We identified two populations of BMDMs unique to GBMs; Mac-1and Mac-2. Mac-1 demonstrates upregulation of immature myeloid gene signature and altered metabolic pathways. Mac-2 is characterized by expression of scavenger receptor MARCO. Pseudotime and RNA velocity analysis revealed the ability of Mac-1 to transition and differentiate to Mac-2 and other GAM subtypes. We further found that the presence of these two populations of BMDMs are associated with the presence of tumor cells with stem cell and mesenchymal features. Bulk RNA-sequencing data demonstrates that gene signatures of these populations are associated with worse survival in GBM. CONCLUSION We used sc-RNAseq to identify a novel population of immature BMDMs that is associated with higher glioma grades. This population exhibited altered metabolic pathways and stem-like potentials to differentiate into other GAM populations including GAMs with upregulation of immunosuppressive pathways. Our results elucidate unique interactions between BMDMs and GBM tumor cells that potentially drives GBM progression and the more aggressive mesenchymal subtype. Our discovery of these novel BMDMs have implications in new therapeutic targets in improving the efficacy of immune-based therapies in GBM.


Sign in / Sign up

Export Citation Format

Share Document