scholarly journals QUBIC2: A novel biclustering algorithm for large-scale bulk RNA-sequencing and single-cell RNA-sequencing data analysis

2018 ◽  
Author(s):  
Juan Xie ◽  
Anjun Ma ◽  
Yu Zhang ◽  
Bingqiang Liu ◽  
Changlin Wan ◽  
...  

ABSTRACTThe combination of biclustering and large-scale gene expression data holds a promising potential for inference of the condition specific functional pathways/networks. However, existing biclustering tools do not have satisfied performance on high-resolution RNA-sequencing (RNA-Seq) data, majorly due to the lack of (i) a consideration of high sparsity of RNA-Seq data, e.g., the massive zeros or lowly expressed genes in the data, especially for single-cell RNA-Seq (scRNA-Seq) data, and (ii) an understanding of the underlying transcriptional regulation signals of the observed gene expression values. Here we presented a novel biclustering algorithm namely QUBIC2, for the analysis of large-scale bulk RNA-Seq and scRNA-Seq data. Key novelties of the algorithm include (i) used a truncated model to handle the unreliable quantification of genes with low or moderate expression, (ii) adopted the mixture Gaussian distribution and an information-divergency objective function to capture shared transcriptional regulation signals among a set of genes, (iii) utilized a Core-Dual strategy to identify biclusters and optimize relevant parameters, and (iv) developed a size-based P-value framework to evaluate the statistical significances of all the identified biclusters. Our method validation on comprehensive data sets of bulk and single cell RNA-seq data suggests that QUBIC2 had superior performance in functional modules detection and cell type classification compared with the other five widely-used biclustering tools. In addition, the applications of temporal and spatial data demonstrated that QUBIC2 can derive meaningful biological information from scRNA-Seq data. The source code for QUBIC2 can be freely accessed at https://github.com/maqin2001/qubic2.

2018 ◽  
Author(s):  
Xianwen Ren ◽  
Liangtao Zheng ◽  
Zemin Zhang

ABSTRACTClustering is a prevalent analytical means to analyze single cell RNA sequencing data but the rapidly expanding data volume can make this process computational challenging. New methods for both accurate and efficient clustering are of pressing needs. Here we proposed a new clustering framework based on random projection and feature construction for large scale single-cell RNA sequencing data, which greatly improves clustering accuracy, robustness and computational efficacy for various state-of-the-art algorithms benchmarked on multiple real datasets. On a dataset with 68,578 human blood cells, our method reached 20% improvements for clustering accuracy and 50-fold acceleration but only consumed 66% memory usage compared to the widely-used software package SC3. Compared to k-means, the accuracy improvement can reach 3-fold depending on the concrete dataset. An R implementation of the framework is available from https://github.com/Japrin/sscClust.


2018 ◽  
Author(s):  
Koen Van Den Berge ◽  
Katharina Hembach ◽  
Charlotte Soneson ◽  
Simone Tiberi ◽  
Lieven Clement ◽  
...  

Gene expression is the fundamental level at which the result of various genetic and regulatory programs are observable. The measurement of transcriptome-wide gene expression has convincingly switched from microarrays to sequencing in a matter of years. RNA sequencing (RNA-seq) provides a quantitative and open system for profiling transcriptional outcomes on a large scale and therefore facilitates a large diversity of applications, including basic science studies, but also agricultural or clinical situations. In the past 10 years or so, much has been learned about the characteristics of the RNA-seq datasets as well as the performance of the myriad of methods developed. In this review, we give an overall view of the developments in RNA-seq data analysis, including experimental design, with an explicit focus on quantification of gene expression and statistical approaches for differential expression. We also highlight emerging data types, such as single-cell RNA-seq and gene expression profiling using long-read technologies.


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.


2022 ◽  
Author(s):  
Sofya Lipnitskaya ◽  
Yang Shen ◽  
Stefan Legewie ◽  
Holger Klein ◽  
Kolja Becker

Abstract Background: Recent studies in the area of transcriptomics performed on single-cell and population levels reveal noticeable variability in gene expression measurements provided by different RNA sequencing technologies. Due to increased noise and complexity of single-cell RNA-Seq (scRNA-Seq) data over the bulk experiment, there is a substantial number of variably-expressed genes and so-called dropouts, challenging the subsequent computational analysis and potentially leading to false positive discoveries. In order to investigate factors affecting technical variability between RNA sequencing experiments of different technologies, we performed a systematic assessment of single-cell and bulk RNA-Seq data, which have undergone the same pre-processing and sample preparation procedures. Results: Our analysis indicates that variability between gene expression measurements as well as dropout events are not exclusively caused by biological variability, low expression levels, or random variation. Furthermore, we propose FAVSeq, a machine learning-assisted pipeline for detection of factors contributing to gene expression variability in matched RNA-Seq data provided by two technologies. Based on the analysis of the matched bulk and single-cell dataset, we found the 3'-UTR and transcript lengths as the most relevant effectors of the observed variation between RNA-Seq experiments, while the same factors together with cellular compartments were shown to be associated with dropouts. Conclusions: Here, we investigated the sources of variation in RNA-Seq profiles of matched single-cell and bulk experiments. In addition, we proposed the FAVSeq pipeline for analyzing multimodal RNA sequencing data, which allowed to identify factors affecting quantitative difference in gene expression measurements as well as the presence of dropouts. Hereby, the derived knowledge can be employed further in order to improve the interpretation of RNA-Seq data and identify genes that can be affected by assay-based deviations. Source code is available under the MIT license at https://github.com/slipnitskaya/FAVSeq.


2020 ◽  
Author(s):  
Lin Li ◽  
Hao Dai ◽  
Zhaoyuan Fang ◽  
Luonan Chen

AbstractThe rapid advancement of single cell technologies has shed new light on the complex mechanisms of cellular heterogeneity. However, compared with bulk RNA sequencing (RNA-seq), single-cell RNA-seq (scRNA-seq) suffers from higher noise and lower coverage, which brings new computational difficulties. Based on statistical independence, cell-specific network (CSN) is able to quantify the overall associations between genes for each cell, yet suffering from a problem of overestimation related to indirect effects. To overcome this problem, we propose the “conditional cell-specific network” (CCSN) method, which can measure the direct associations between genes by eliminating the indirect associations. CCSN can be used for cell clustering and dimension reduction on a network basis of single cells. Intuitively, each CCSN can be viewed as the transformation from less “reliable” gene expression to more “reliable” gene-gene associations in a cell. Based on CCSN, we further design network flow entropy (NFE) to estimate the differentiation potency of a single cell. A number of scRNA-seq datasets were used to demonstrate the advantages of our approach: (1) one direct association network for one cell; (2) most existing scRNA-seq methods designed for gene expression matrices are also applicable to CCSN-transformed degree matrices; (3) CCSN-based NFE helps resolving the direction of differentiation trajectories by quantifying the potency of each cell. CCSN is publicly available at http://sysbio.sibcb.ac.cn/cb/chenlab/soft/CCSN.zip.


2021 ◽  
Vol 17 (5) ◽  
pp. e1009029
Author(s):  
Weimiao Wu ◽  
Yunqing Liu ◽  
Qile Dai ◽  
Xiting Yan ◽  
Zuoheng Wang

Single-cell RNA sequencing technology provides an opportunity to study gene expression at single-cell resolution. However, prevalent dropout events result in high data sparsity and noise that may obscure downstream analyses in single-cell transcriptomic studies. We propose a new method, G2S3, that imputes dropouts by borrowing information from adjacent genes in a sparse gene graph learned from gene expression profiles across cells. We applied G2S3 and ten existing imputation methods to eight single-cell transcriptomic datasets and compared their performance. Our results demonstrated that G2S3 has superior overall performance in recovering gene expression, identifying cell subtypes, reconstructing cell trajectories, identifying differentially expressed genes, and recovering gene regulatory and correlation relationships. Moreover, G2S3 is computationally efficient for imputation in large-scale single-cell transcriptomic datasets.


Author(s):  
Koen Van Den Berge ◽  
Katharina Hembach ◽  
Charlotte Soneson ◽  
Simone Tiberi ◽  
Lieven Clement ◽  
...  

Gene expression is the fundamental level at which the result of various genetic and regulatory programs are observable. The measurement of transcriptome-wide gene expression has convincingly switched from microarrays to sequencing in a matter of years. RNA sequencing (RNA-seq) provides a quantitative and open system for profiling transcriptional outcomes on a large scale and therefore facilitates a large diversity of applications, including basic science studies, but also agricultural or clinical situations. In the past 10 years or so, much has been learned about the characteristics of the RNA-seq datasets as well as the performance of the myriad of methods developed. In this review, we give an overall view of the developments in RNA-seq data analysis, including experimental design, with an explicit focus on quantification of gene expression and statistical approaches for differential expression. We also highlight emerging data types, such as single-cell RNA-seq and gene expression profiling using long-read technologies.


2019 ◽  
Vol 2 (1) ◽  
pp. 139-173 ◽  
Author(s):  
Koen Van den Berge ◽  
Katharina M. Hembach ◽  
Charlotte Soneson ◽  
Simone Tiberi ◽  
Lieven Clement ◽  
...  

Gene expression is the fundamental level at which the results of various genetic and regulatory programs are observable. The measurement of transcriptome-wide gene expression has convincingly switched from microarrays to sequencing in a matter of years. RNA sequencing (RNA-seq) provides a quantitative and open system for profiling transcriptional outcomes on a large scale and therefore facilitates a large diversity of applications, including basic science studies, but also agricultural or clinical situations. In the past 10 years or so, much has been learned about the characteristics of the RNA-seq data sets, as well as the performance of the myriad of methods developed. In this review, we give an overview of the developments in RNA-seq data analysis, including experimental design, with an explicit focus on the quantification of gene expression and statistical approachesfor differential expression. We also highlight emerging data types, such as single-cell RNA-seq and gene expression profiling using long-read technologies.


Author(s):  
Koen Van Den Berge ◽  
Katharina Hembach ◽  
Charlotte Soneson ◽  
Simone Tiberi ◽  
Lieven Clement ◽  
...  

Gene expression is the fundamental level at which the result of various genetic and regulatory programs are observable. The measurement of transcriptome-wide gene expression has convincingly switched from microarrays to sequencing in a matter of years. RNA sequencing (RNA-seq) provides a quantitative and open system for profiling transcriptional outcomes on a large scale and therefore facilitates a large diversity of applications, including basic science studies, but also agricultural or clinical situations. In the past 10 years or so, much has been learned about the characteristics of the RNA-seq datasets as well as the performance of the myriad of methods developed. In this review, we give an overall view of the developments in RNA-seq data analysis, including experimental design, with an explicit focus on quantification of gene expression and statistical approaches for differential expression. We also highlight emerging data types, such as single-cell RNA-seq and gene expression profiling using long-read technologies.


2022 ◽  
Author(s):  
Sofya Lipnitskaya ◽  
Yang Shen ◽  
Stefan Legewie ◽  
Holger Klein ◽  
Kolja Becker

Background: Recent studies in the area of transcriptomics performed on single-cell and population levels reveal noticeable variability in gene expression measurements provided by different RNA sequencing technologies. Due to increased noise and complexity of single-cell RNA-Seq (scRNA-Seq) data over the bulk experiment, there is a substantial number of variably-expressed genes and so-called dropouts, challenging the subsequent computational analysis and potentially leading to false positive discoveries. In order to investigate factors affecting technical variability between RNA sequencing experiments of different technologies, we performed a systematic assessment of single-cell and bulk RNA-Seq data, which have undergone the same pre-processing and sample preparation procedures. Results: Our analysis indicates that variability between gene expression measurements as well as dropout events are not exclusively caused by biological variability, low expression levels, or random variation. Furthermore, we propose FAVSeq, a machine learning-assisted pipeline for detection of factors contributing to gene expression variability in matched RNA-Seq data provided by two technologies. Based on the analysis of the matched bulk and single-cell dataset, we found the 3'-UTR and transcript lengths as the most relevant effectors of the observed variation between RNA-Seq experiments, while the same factors together with cellular compartments were shown to be associated with dropouts. Conclusions: Here, we investigated the sources of variation in RNA-Seq profiles of matched single-cell and bulk experiments. In addition, we proposed the FAVSeq pipeline for analyzing multimodal RNA sequencing data, which allowed to identify factors affecting quantitative difference in gene expression measurements as well as the presence of dropouts. Hereby, the derived knowledge can be employed further in order to improve the interpretation of RNA-Seq data and identify genes that can be affected by assay-based deviations. Source code is available under the MIT license at https://github.com/slipnitskaya/FAVSeq.


Sign in / Sign up

Export Citation Format

Share Document