scholarly journals scImpute: Accurate And Robust Imputation For Single Cell RNA-Seq Data

2017 ◽  
Author(s):  
Wei Vivian Li ◽  
Jingyi Jessica Li

The emerging single cell RNA sequencing (scRNA-seq) technologies enable the investigation of transcriptomic landscapes at single-cell resolution. The analysis of scRNA-seq data is complicated by excess zero or near zero counts, the so-called dropouts due to the low amounts of mRNA sequenced within individual cells. Downstream analysis of scRNA-seq would be severely biased if the dropout events are not properly corrected. We introduce scImpute, a statistical method to accurately and robustly impute the dropout values in scRNA-seq data. ScImpute automatically identifies gene expression values affected by dropout events, and only perform imputation on these values without introducing new bias to the rest data. ScImpute also detects outlier or rare cells and excludes them from imputation. Evaluation based on both simulated and real scRNA-seq data on mouse embryos, mouse brain cells, human blood cells, and human embryonic stem cells suggests that scImpute is an effective tool to recover transcriptome dynamics masked by dropout events. scImpute is shown to correct false zero counts, enhance the clustering of cell populations and subpopulations, improve the accuracy of differential expression analysis, and aid the study of gene expression dynamics.

2021 ◽  
Vol 17 (6) ◽  
pp. e1009118
Author(s):  
Jing Qi ◽  
Yang Zhou ◽  
Zicen Zhao ◽  
Shuilin Jin

The single-cell RNA sequencing (scRNA-seq) technologies obtain gene expression at single-cell resolution and provide a tool for exploring cell heterogeneity and cell types. As the low amount of extracted mRNA copies per cell, scRNA-seq data exhibit a large number of dropouts, which hinders the downstream analysis of the scRNA-seq data. We propose a statistical method, SDImpute (Single-cell RNA-seq Dropout Imputation), to implement block imputation for dropout events in scRNA-seq data. SDImpute automatically identifies the dropout events based on the gene expression levels and the variations of gene expression across similar cells and similar genes, and it implements block imputation for dropouts by utilizing gene expression unaffected by dropouts from similar cells. In the experiments, the results of the simulated datasets and real datasets suggest that SDImpute is an effective tool to recover the data and preserve the heterogeneity of gene expression across cells. Compared with the state-of-the-art imputation methods, SDImpute improves the accuracy of the downstream analysis including clustering, visualization, and differential expression analysis.


2019 ◽  
Author(s):  
Marcus Alvarez ◽  
Elior Rahmani ◽  
Brandon Jew ◽  
Kristina M. Garske ◽  
Zong Miao ◽  
...  

AbstractSingle-nucleus RNA sequencing (snRNA-seq) measures gene expression in individual nuclei instead of cells, allowing for unbiased cell type characterization in solid tissues. Contrary to single-cell RNA seq (scRNA-seq), we observe that snRNA-seq is commonly subject to contamination by high amounts of extranuclear background RNA, which can lead to identification of spurious cell types in downstream clustering analyses if overlooked. We present a novel approach to remove debris-contaminated droplets in snRNA-seq experiments, called Debris Identification using Expectation Maximization (DIEM). Our likelihood-based approach models the gene expression distribution of debris and cell types, which are estimated using EM. We evaluated DIEM using three snRNA-seq data sets: 1) human differentiating preadipocytes in vitro, 2) fresh mouse brain tissue, and 3) human frozen adipose tissue (AT) from six individuals. All three data sets showed various degrees of extranuclear RNA contamination. We observed that existing methods fail to account for contaminated droplets and led to spurious cell types. When compared to filtering using these state of the art methods, DIEM better removed droplets containing high levels of extranuclear RNA and led to higher quality clusters. Although DIEM was designed for snRNA-seq data, we also successfully applied DIEM to single-cell data. To conclude, our novel method DIEM removes debris-contaminated droplets from single-cell-based data fast and effectively, leading to cleaner downstream analysis. Our code is freely available for use at https://github.com/marcalva/diem.


2018 ◽  
Vol 20 (4) ◽  
pp. 1583-1589 ◽  
Author(s):  
Shun H Yip ◽  
Pak Chung Sham ◽  
Junwen Wang

Abstract Traditional RNA sequencing (RNA-seq) allows the detection of gene expression variations between two or more cell populations through differentially expressed gene (DEG) analysis. However, genes that contribute to cell-to-cell differences are not discoverable with RNA-seq because RNA-seq samples are obtained from a mixture of cells. Single-cell RNA-seq (scRNA-seq) allows the detection of gene expression in each cell. With scRNA-seq, highly variable gene (HVG) discovery allows the detection of genes that contribute strongly to cell-to-cell variation within a homogeneous cell population, such as a population of embryonic stem cells. This analysis is implemented in many software packages. In this study, we compare seven HVG methods from six software packages, including BASiCS, Brennecke, scLVM, scran, scVEGs and Seurat. Our results demonstrate that reproducibility in HVG analysis requires a larger sample size than DEG analysis. Discrepancies between methods and potential issues in these tools are discussed and recommendations are made.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Giancarlo Bonora ◽  
Vijay Ramani ◽  
Ritambhara Singh ◽  
He Fang ◽  
Dana L. Jackson ◽  
...  

Abstract Background Mammalian development is associated with extensive changes in gene expression, chromatin accessibility, and nuclear structure. Here, we follow such changes associated with mouse embryonic stem cell differentiation and X inactivation by integrating, for the first time, allele-specific data from these three modalities obtained by high-throughput single-cell RNA-seq, ATAC-seq, and Hi-C. Results Allele-specific contact decay profiles obtained by single-cell Hi-C clearly show that the inactive X chromosome has a unique profile in differentiated cells that have undergone X inactivation. Loss of this inactive X-specific structure at mitosis is followed by its reappearance during the cell cycle, suggesting a “bookmark” mechanism. Differentiation of embryonic stem cells to follow the onset of X inactivation is associated with changes in contact decay profiles that occur in parallel on both the X chromosomes and autosomes. Single-cell RNA-seq and ATAC-seq show evidence of a delay in female versus male cells, due to the presence of two active X chromosomes at early stages of differentiation. The onset of the inactive X-specific structure in single cells occurs later than gene silencing, consistent with the idea that chromatin compaction is a late event of X inactivation. Single-cell Hi-C highlights evidence of discrete changes in nuclear structure characterized by the acquisition of very long-range contacts throughout the nucleus. Novel computational approaches allow for the effective alignment of single-cell gene expression, chromatin accessibility, and 3D chromosome structure. Conclusions Based on trajectory analyses, three distinct nuclear structure states are detected reflecting discrete and profound simultaneous changes not only to the structure of the X chromosomes, but also to that of autosomes during differentiation. Our study reveals that long-range structural changes to chromosomes appear as discrete events, unlike progressive changes in gene expression and chromatin accessibility.


2021 ◽  
Author(s):  
Hanbyeol Kim ◽  
Joongho Lee ◽  
Keunsoo Kang ◽  
Seokhyun Yoon

Abstract Cell type identification is a key step to downstream analysis of single cell RNA-seq experiments. Indispensible information for this is gene expression, which is used to cluster cells, train the model and set rejection thresholds. Problem is they are subject to batch effect arising from different platforms and preprocessing. We present MarkerCount, which uses the number of markers expressed regardless of their expression level to initially identify cell types and, then, reassign cell type in cluster-basis. MarkerCount works both in reference and marker-based mode, where the latter utilizes only the existing lists of markers, while the former required pre-annotated dataset to train the model. The performance was evaluated and compared with the existing identifiers, both marker and reference-based, that can be customized with publicly available datasets and marker DB. The results show that MarkerCount provides a stable performance when comparing with other reference-based and marker-based cell type identifiers.


2017 ◽  
Author(s):  
Koen Van den Berge ◽  
Charlotte Soneson ◽  
Michael I. Love ◽  
Mark D. Robinson ◽  
Lieven Clement

AbstractDropout in single cell RNA-seq (scRNA-seq) applications causes many transcripts to go undetected. It induces excess zero counts, which leads to power issues in differential expression (DE) analysis and has triggered the development of bespoke scRNA-seq DE tools that cope with zero-inflation. Recent evaluations, however, have shown that dedicated scRNA-seq tools provide no advantage compared to traditional bulk RNA-seq tools. We introduce zingeR, a zero-inflated negative binomial model that identifies excess zero counts and generates observation weights to unlock bulk RNA-seq pipelines for zero-inflation, boosting performance in scRNA-seq differential expression analysis.


2020 ◽  
Vol 36 (10) ◽  
pp. 3131-3138
Author(s):  
Ke Jin ◽  
Le Ou-Yang ◽  
Xing-Ming Zhao ◽  
Hong Yan ◽  
Xiao-Fei Zhang

Abstract Motivation Single-cell RNA sequencing (scRNA-seq) methods make it possible to reveal gene expression patterns at single-cell resolution. Due to technical defects, dropout events in scRNA-seq will add noise to the gene-cell expression matrix and hinder downstream analysis. Therefore, it is important for recovering the true gene expression levels before carrying out downstream analysis. Results In this article, we develop an imputation method, called scTSSR, to recover gene expression for scRNA-seq. Unlike most existing methods that impute dropout events by borrowing information across only genes or cells, scTSSR simultaneously leverages information from both similar genes and similar cells using a two-side sparse self-representation model. We demonstrate that scTSSR can effectively capture the Gini coefficients of genes and gene-to-gene correlations observed in single-molecule RNA fluorescence in situ hybridization (smRNA FISH). Down-sampling experiments indicate that scTSSR performs better than existing methods in recovering the true gene expression levels. We also show that scTSSR has a competitive performance in differential expression analysis, cell clustering and cell trajectory inference. Availability and implementation The R package is available at https://github.com/Zhangxf-ccnu/scTSSR. Supplementary information Supplementary data are available at Bioinformatics online.


2019 ◽  
Author(s):  
Reiichi Sugihara ◽  
Yuki Kato ◽  
Tomoya Mori ◽  
Yukio Kawahara

AbstractRecent techniques on single-cell RNA sequencing have boosted transcriptome-wide observation of gene expression dynamics of time-course data at a single-cell scale. Typical examples of such analysis include inference of a pseudotime cell trajectory, and comparison of pseudotime trajectories between different experimental conditions will tell us how feature genes regulate a dynamic cellular process. Existing methods for comparing pseudotime trajectories, however, force users to select trajectories to be compared because they can deal only with simple linear trajectories, leading to the possibility of making a biased interpretation. Here we present CAPITAL, a method for comparing pseudotime trajectories with tree alignment whereby trajectories including branching can be compared without any knowledge of paths to be compared. Computational tests on time-series public data indicate that CAPITAL can align non-linear pseudotime trajectories and reveal gene expression dynamics.


Genes ◽  
2020 ◽  
Vol 11 (5) ◽  
pp. 532 ◽  
Author(s):  
Weilai Chi ◽  
Minghua Deng

Single-cell RNA-seq (scRNA-seq) is quite prevalent in studying transcriptomes, but it suffers from excessive zeros, some of which are true, but others are false. False zeros, which can be seen as missing data, obstruct the downstream analysis of single-cell RNA-seq data. How to distinguish true zeros from false ones is the key point of this problem. Here, we propose sparsity-penalized stacked denoising autoencoders (scSDAEs) to impute scRNA-seq data. scSDAEs adopt stacked denoising autoencoders with a sparsity penalty, as well as a layer-wise pretraining procedure to improve model fitting. scSDAEs can capture nonlinear relationships among the data and incorporate information about the observed zeros. We tested the imputation efficiency of scSDAEs on recovering the true values of gene expression and helping downstream analysis. First, we show that scSDAE can recover the true values and the sample–sample correlations of bulk sequencing data with simulated noise. Next, we demonstrate that scSDAEs accurately impute RNA mixture dataset with different dilutions, spike-in RNA concentrations affected by technical zeros, and improves the consistency of RNA and protein levels in CITE-seq data. Finally, we show that scSDAEs can help downstream clustering analysis. In this study, we develop a deep learning-based method, scSDAE, to impute single-cell RNA-seq affected by technical zeros. Furthermore, we show that scSDAEs can recover the true values, to some extent, and help downstream analysis.


Sign in / Sign up

Export Citation Format

Share Document