scholarly journals PROSSTT: probabilistic simulation of single-cell RNA-seq data for complex differentiation processes

2019 ◽  
Vol 35 (18) ◽  
pp. 3517-3519 ◽  
Author(s):  
Nikolaos Papadopoulos ◽  
Parra R Gonzalo ◽  
Johannes Söding

Abstract Summary Cellular lineage trees can be derived from single-cell RNA sequencing snapshots of differentiating cells. Currently, only datasets with simple topologies are available. To test and further develop tools for lineage tree reconstruction, we need test datasets with known complex topologies. PROSSTT can simulate scRNA-seq datasets for differentiation processes with lineage trees of any desired complexity, noise level, noise model and size. PROSSTT also provides scripts to quantify the quality of predicted lineage trees. Availability and implementation https://github.com/soedinglab/prosstt. Supplementary information Supplementary data are available at Bioinformatics online.

2018 ◽  
Author(s):  
Nikolaos Papadopoulos ◽  
R. Gonzalo Parra ◽  
Johannes Söding

BackgroundSingle-cell RNA sequencing (scRNA-seq) is an enabling technology for the study of cellular differentiation and heterogeneity. From snapshots of the transcriptomic profiles of differentiating single cells, the cellular lineage tree that leads from a progenitor population to multiple types of differentiated cells can be derived. The underlying lineage trees of most published datasets are linear or have a single branchpoint, but many studies with more complex lineage trees will soon become available. To test and further develop tools for lineage tree reconstruction, we need test datasets with known trees.ResultsPROSSTT can simulate scRNA-seq datasets for differentiation processes with lineage trees of any desired complexity, noise level, noise model, and size. PROSSTT also provides scripts to quantify the quality of predicted lineage trees.Availabilityhttps://github.com/soedinglab/[email protected]


2019 ◽  
Author(s):  
Alemu Takele Assefa ◽  
Jo Vandesompele ◽  
Olivier Thas

SummarySPsimSeq is a semi-parametric simulation method for bulk and single cell RNA sequencing data. It simulates data from a good estimate of the actual distribution of a given real RNA-seq dataset. In contrast to existing approaches that assume a particular data distribution, our method constructs an empirical distribution of gene expression data from a given source RNA-seq experiment to faithfully capture the data characteristics of real data. Importantly, our method can be used to simulate a wide range of scenarios, such as single or multiple biological groups, systematic variations (e.g. confounding batch effects), and different sample sizes. It can also be used to simulate different gene expression units resulting from different library preparation protocols, such as read counts or UMI counts.Availability and implementationThe R package and associated documentation is available from https://github.com/CenterForStatistics-UGent/SPsimSeq.Supplementary informationSupplementary data are available at bioRχiv online.


2021 ◽  
Author(s):  
Federico Agostinis ◽  
Chiara Romualdi ◽  
Gabriele Sales ◽  
Davide Risso

Summary: We present NewWave, a scalable R/Bioconductor package for the dimensionality reduction and batch effect removal of single-cell RNA sequencing data. To achieve scalability, NewWave uses mini-batch optimization and can work with out-of-memory data, enabling users to analyze datasets with millions of cells. Availability and implementation: NewWave is implemented as an open-source R package available through the Bioconductor project at https://bioconductor.org/packages/NewWave/ Supplementary information: Supplementary data are available at Bioinformatics online.


2020 ◽  
Vol 36 (19) ◽  
pp. 4860-4868
Author(s):  
Kenong Su ◽  
Zhijin Wu ◽  
Hao Wu

Abstract Motivation Determining the sample size for adequate power to detect statistical significance is a crucial step at the design stage for high-throughput experiments. Even though a number of methods and tools are available for sample size calculation for microarray and RNA-seq in the context of differential expression (DE), this topic in the field of single-cell RNA sequencing is understudied. Moreover, the unique data characteristics present in scRNA-seq such as sparsity and heterogeneity increase the challenge. Results We propose POWSC, a simulation-based method, to provide power evaluation and sample size recommendation for single-cell RNA-sequencing DE analysis. POWSC consists of a data simulator that creates realistic expression data, and a power assessor that provides a comprehensive evaluation and visualization of the power and sample size relationship. The data simulator in POWSC outperforms two other state-of-art simulators in capturing key characteristics of real datasets. The power assessor in POWSC provides a variety of power evaluations including stratified and marginal power analyses for DEs characterized by two forms (phase transition or magnitude tuning), under different comparison scenarios. In addition, POWSC offers information for optimizing the tradeoffs between sample size and sequencing depth with the same total reads. Availability and implementation POWSC is an open-source R package available online at https://github.com/suke18/POWSC. Supplementary information Supplementary data are available at Bioinformatics online.


2020 ◽  
Vol 36 (11) ◽  
pp. 3588-3589 ◽  
Author(s):  
Kaiyi Zhu ◽  
Dimitris Anastassiou

Abstract Summary We developed 2DImpute, an imputation method for correcting false zeros (known as dropouts) in single-cell RNA-sequencing (scRNA-seq) data. It features preventing excessive correction by predicting the false zeros and imputing their values by making use of the interrelationships between both genes and cells in the expression matrix. We showed that 2DImpute outperforms several leading imputation methods by applying it on datasets from various scRNA-seq protocols. Availability and implementation The R package of 2DImpute is freely available at GitHub (https://github.com/zky0708/2DImpute). Contact [email protected] Supplementary information Supplementary data are available at Bioinformatics online.


Author(s):  
Irzam Sarfraz ◽  
Muhammad Asif ◽  
Joshua D Campbell

Abstract Motivation R Experiment objects such as the SummarizedExperiment or SingleCellExperiment are data containers for storing one or more matrix-like assays along with associated row and column data. These objects have been used to facilitate the storage and analysis of high-throughput genomic data generated from technologies such as single-cell RNA sequencing. One common computational task in many genomics analysis workflows is to perform subsetting of the data matrix before applying down-stream analytical methods. For example, one may need to subset the columns of the assay matrix to exclude poor-quality samples or subset the rows of the matrix to select the most variable features. Traditionally, a second object is created that contains the desired subset of assay from the original object. However, this approach is inefficient as it requires the creation of an additional object containing a copy of the original assay and leads to challenges with data provenance. Results To overcome these challenges, we developed an R package called ExperimentSubset, which is a data container that implements classes for efficient storage and streamlined retrieval of assays that have been subsetted by rows and/or columns. These classes are able to inherently provide data provenance by maintaining the relationship between the subsetted and parent assays. We demonstrate the utility of this package on a single-cell RNA-seq dataset by storing and retrieving subsets at different stages of the analysis while maintaining a lower memory footprint. Overall, the ExperimentSubset is a flexible container for the efficient management of subsets. Availability and implementation ExperimentSubset package is available at Bioconductor: https://bioconductor.org/packages/ExperimentSubset/ and Github: https://github.com/campbio/ExperimentSubset. Supplementary information Supplementary data are available at Bioinformatics online.


Author(s):  
Yang Xu ◽  
Priyojit Das ◽  
Rachel Patton McCord

Abstract Motivation Deep learning approaches have empowered single-cell omics data analysis in many ways and generated new insights from complex cellular systems. As there is an increasing need for single cell omics data to be integrated across sources, types, and features of data, the challenges of integrating single-cell omics data are rising. Here, we present an unsupervised deep learning algorithm that learns discriminative representations for single-cell data via maximizing mutual information, SMILE (Single-cell Mutual Information Learning). Results Using a unique cell-pairing design, SMILE successfully integrates multi-source single-cell transcriptome data, removing batch effects and projecting similar cell types, even from different tissues, into the shared space. SMILE can also integrate data from two or more modalities, such as joint profiling technologies using single-cell ATAC-seq, RNA-seq, DNA methylation, Hi-C, and ChIP data. When paired cells are known, SMILE can integrate data with unmatched feature, such as genes for RNA-seq and genome wide peaks for ATAC-seq. Integrated representations learned from joint profiling technologies can then be used as a framework for comparing independent single source data. Supplementary information Supplementary data are available at Bioinformatics online. The source code of SMILE including analyses of key results in the study can be found at: https://github.com/rpmccordlab/SMILE.


2017 ◽  
Author(s):  
Zhun Miao ◽  
Ke Deng ◽  
Xiaowo Wang ◽  
Xuegong Zhang

AbstractSummaryThe excessive amount of zeros in single-cell RNA-seq data include “real” zeros due to the on-off nature of gene transcription in single cells and “dropout” zeros due to technical reasons. Existing differential expression (DE) analysis methods cannot distinguish these two types of zeros. We developed an R package DEsingle which employed Zero-Inflated Negative Binomial model to estimate the proportion of real and dropout zeros and to define and detect 3 types of DE genes in single-cell RNA-seq data with higher accuracy.Availability and ImplementationThe R package DEsingle is freely available at https://github.com/miaozhun/DEsingle and is under Bioconductor’s consideration [email protected] informationSupplementary data are available at bioRxiv online.


2017 ◽  
Author(s):  
Gioele La Manno ◽  
Ruslan Soldatov ◽  
Hannah Hochgerner ◽  
Amit Zeisel ◽  
Viktor Petukhov ◽  
...  

AbstractRNA abundance is a powerful indicator of the state of individual cells, but does not directly reveal dynamic processes such as cellular differentiation. Here we show that RNA velocity—the time derivative of RNA abundance—can be estimated by distinguishing unspliced and spliced mRNAs in standard single-cell RNA sequencing protocols. We show that RNA velocity is a vector that predicts the future state of individual cells on a timescale of hours. We validate the accuracy of RNA velocity in the neural crest lineage, demonstrate its use on multiple technical platforms, reconstruct the branching lineage tree of the mouse hippocampus, and measure RNA kinetics in human embryonic brain. We expect RNA velocity to greatly aid the analysis of developmental lineages and cellular dynamics, particularly in humans.


2019 ◽  
Vol 35 (24) ◽  
pp. 5155-5162 ◽  
Author(s):  
Chengzhong Ye ◽  
Terence P Speed ◽  
Agus Salim

Abstract Motivation Dropout is a common phenomenon in single-cell RNA-seq (scRNA-seq) data, and when left unaddressed it affects the validity of the statistical analyses. Despite this, few current methods for differential expression (DE) analysis of scRNA-seq data explicitly model the process that gives rise to the dropout events. We develop DECENT, a method for DE analysis of scRNA-seq data that explicitly and accurately models the molecule capture process in scRNA-seq experiments. Results We show that DECENT demonstrates improved DE performance over existing DE methods that do not explicitly model dropout. This improvement is consistently observed across several public scRNA-seq datasets generated using different technological platforms. The gain in improvement is especially large when the capture process is overdispersed. DECENT maintains type I error well while achieving better sensitivity. Its performance without spike-ins is almost as good as when spike-ins are used to calibrate the capture model. Availability and implementation The method is implemented as a publicly available R package available from https://github.com/cz-ye/DECENT. Supplementary information Supplementary data are available at Bioinformatics online.


Sign in / Sign up

Export Citation Format

Share Document