scholarly journals Integration of single-cell and bulk RNA sequencing data reveals key cell types and regulators in traumatic brain injury

2021 ◽  
Vol 18 (2) ◽  
pp. 1201-1214
Author(s):  
Rui-zhe Zheng ◽  
◽  
Jin Xing ◽  
Qiong Huang ◽  
Xi-tao Yang ◽  
...  
Author(s):  
Yinlei Hu ◽  
Bin Li ◽  
Falai Chen ◽  
Kun Qu

Abstract Unsupervised clustering is a fundamental step of single-cell RNA sequencing data analysis. This issue has inspired several clustering methods to classify cells in single-cell RNA sequencing data. However, accurate prediction of the cell clusters remains a substantial challenge. In this study, we propose a new algorithm for single-cell RNA sequencing data clustering based on Sparse Optimization and low-rank matrix factorization (scSO). We applied our scSO algorithm to analyze multiple benchmark datasets and showed that the cluster number predicted by scSO was close to the number of reference cell types and that most cells were correctly classified. Our scSO algorithm is available at https://github.com/QuKunLab/scSO. Overall, this study demonstrates a potent cell clustering approach that can help researchers distinguish cell types in single-cell RNA sequencing data.


2019 ◽  
Vol 21 (5) ◽  
pp. 1581-1595 ◽  
Author(s):  
Xinlei Zhao ◽  
Shuang Wu ◽  
Nan Fang ◽  
Xiao Sun ◽  
Jue Fan

Abstract Single-cell RNA sequencing (scRNA-seq) has been rapidly developing and widely applied in biological and medical research. Identification of cell types in scRNA-seq data sets is an essential step before in-depth investigations of their functional and pathological roles. However, the conventional workflow based on clustering and marker genes is not scalable for an increasingly large number of scRNA-seq data sets due to complicated procedures and manual annotation. Therefore, a number of tools have been developed recently to predict cell types in new data sets using reference data sets. These methods have not been generally adapted due to a lack of tool benchmarking and user guidance. In this article, we performed a comprehensive and impartial evaluation of nine classification software tools specifically designed for scRNA-seq data sets. Results showed that Seurat based on random forest, SingleR based on correlation analysis and CaSTLe based on XGBoost performed better than others. A simple ensemble voting of all tools can improve the predictive accuracy. Under nonideal situations, such as small-sized and class-imbalanced reference data sets, tools based on cluster-level similarities have superior performance. However, even with the function of assigning ‘unassigned’ labels, it is still challenging to catch novel cell types by solely using any of the single-cell classifiers. This article provides a guideline for researchers to select and apply suitable classification tools in their analysis workflows and sheds some lights on potential direction of future improvement on classification tools.


2017 ◽  
Author(s):  
Luke Zappia ◽  
Belinda Phipson ◽  
Alicia Oshlack

AbstractAs single-cell RNA sequencing technologies have rapidly developed, so have analysis methods. Many methods have been tested, developed and validated using simulated datasets. Unfortunately, current simulations are often poorly documented, their similarity to real data is not demonstrated, or reproducible code is not available.Here we present the Splatter Bioconductor package for simple, reproducible and well-documented simulation of single-cell RNA-seq data. Splatter provides an interface to multiple simulation methods including Splat, our own simulation, based on a gamma-Poisson distribution. Splat can simulate single populations of cells, populations with multiple cell types or differentiation paths.


2021 ◽  
Author(s):  
Saptarshi Bej ◽  
Anne-Marie Galow ◽  
Robert David ◽  
Markus Wolfien ◽  
Olaf Wolkenhauer

AbstractThe research landscape of single-cell and single-nuclei RNA sequencing is evolving rapidly, and one area that is enabled by this technology, is the detection of rare cells. An automated, unbiased and accurate annotation of rare subpopulations is challenging. Once rare cells are identified in one dataset, it will usually be necessary to generate other datasets to enrich the analysis (e.g., with samples from other tissues). From a machine learning perspective, the challenge arises from the fact that rare cell subpopulations constitute an imbalanced classification problem.We here introduce a Machine Learning (ML)-based oversampling method that uses gene expression counts of already identified rare cells as an input to generate synthetic cells to then identify similar (rare) cells in other publicly available experiments. We utilize single-cell synthetic oversampling (sc-SynO), which is based on the Localized Random Affine Shadowsampling (LoRAS) algorithm. The algorithm corrects for the overall imbalance ratio of the minority and majority class.We demonstrate the effectiveness of the method for two independent use cases, each consisting of two published datasets. The first use case identifies cardiac glial cells in snRNA-Seq data (17 nuclei out of 8,635). This use case was designed to take a larger imbalance ratio (∼1 to 500) into account and only uses single-nuclei data. The second use case was designed to jointly use snRNA-Seq data and scRNA-Seq on a lower imbalance ratio (∼1 to 26) for the training step to likewise investigate the potential of the algorithm to consider both single cell capture procedures and the impact of “less” rare-cell types. For validation purposes, all datasets have also been analyzed in a traditional manner using common data analysis approaches, such as the Seurat3 workflow.Our algorithm identifies rare-cell populations with a high accuracy and low false positive detection rate. A striking benefit of our algorithm is that it can be readily implemented in other and existing workflows. The code basis is publicly available at FairdomHub (https://fairdomhub.org/assays/1368) and can easily be transferred to train other customized approaches.


eLife ◽  
2020 ◽  
Vol 9 ◽  
Author(s):  
MGP van der Wijst ◽  
DH de Vries ◽  
HE Groot ◽  
G Trynka ◽  
CC Hon ◽  
...  

In recent years, functional genomics approaches combining genetic information with bulk RNA-sequencing data have identified the downstream expression effects of disease-associated genetic risk factors through so-called expression quantitative trait locus (eQTL) analysis. Single-cell RNA-sequencing creates enormous opportunities for mapping eQTLs across different cell types and in dynamic processes, many of which are obscured when using bulk methods. Rapid increase in throughput and reduction in cost per cell now allow this technology to be applied to large-scale population genetics studies. To fully leverage these emerging data resources, we have founded the single-cell eQTLGen consortium (sc-eQTLGen), aimed at pinpointing the cellular contexts in which disease-causing genetic variants affect gene expression. Here, we outline the goals, approach and potential utility of the sc-eQTLGen consortium. We also provide a set of study design considerations for future single-cell eQTL studies.


2018 ◽  
Author(s):  
Aaron T. L. Lun ◽  
Samantha Riesenfeld ◽  
Tallulah Andrews ◽  
Tomas Gomes ◽  
John C. Marioni ◽  
...  

AbstractDroplet-based single-cell RNA sequencing protocols have dramatically increased the throughput and efficiency of single-cell transcriptomics studies. A key computational challenge when processing these data is to distinguish libraries for real cells from empty droplets. Existing methods for cell calling set a minimum threshold on the total unique molecular identifier (UMI) count for each library, which indiscriminately discards cell libraries with low UMI counts. Here, we describe a new statistical method for calling cells from droplet-based data, based on detecting significant deviations from the expression profile of the ambient solution. Using simulations, we demonstrate that our method has greater power than existing approaches for detecting cell libraries with low UMI counts, while controlling the false discovery rate among detected cells. We also apply our method to real data, where we show that the use of our method results in the retention of distinct cell types that would otherwise have been discarded.


GigaScience ◽  
2019 ◽  
Vol 8 (10) ◽  
Author(s):  
Yun-Ching Chen ◽  
Abhilash Suresh ◽  
Chingiz Underbayev ◽  
Clare Sun ◽  
Komudi Singh ◽  
...  

AbstractBackgroundIn single-cell RNA-sequencing analysis, clustering cells into groups and differentiating cell groups by differentially expressed (DE) genes are 2 separate steps for investigating cell identity. However, the ability to differentiate between cell groups could be affected by clustering. This interdependency often creates a bottleneck in the analysis pipeline, requiring researchers to repeat these 2 steps multiple times by setting different clustering parameters to identify a set of cell groups that are more differentiated and biologically relevant.FindingsTo accelerate this process, we have developed IKAP—an algorithm to identify major cell groups and improve differentiating cell groups by systematically tuning parameters for clustering. We demonstrate that, with default parameters, IKAP successfully identifies major cell types such as T cells, B cells, natural killer cells, and monocytes in 2 peripheral blood mononuclear cell datasets and recovers major cell types in a previously published mouse cortex dataset. These major cell groups identified by IKAP present more distinguishing DE genes compared with cell groups generated by different combinations of clustering parameters. We further show that cell subtypes can be identified by recursively applying IKAP within identified major cell types, thereby delineating cell identities in a multi-layered ontology.ConclusionsBy tuning the clustering parameters to identify major cell groups, IKAP greatly improves the automation of single-cell RNA-sequencing analysis to produce distinguishing DE genes and refine cell ontology using single-cell RNA-sequencing data.


2021 ◽  
Vol 22 (1) ◽  
Author(s):  
Bobby Ranjan ◽  
Florian Schmidt ◽  
Wenjie Sun ◽  
Jinyu Park ◽  
Mohammad Amin Honardoost ◽  
...  

Abstract Background Clustering is a crucial step in the analysis of single-cell data. Clusters identified in an unsupervised manner are typically annotated to cell types based on differentially expressed genes. In contrast, supervised methods use a reference panel of labelled transcriptomes to guide both clustering and cell type identification. Supervised and unsupervised clustering approaches have their distinct advantages and limitations. Therefore, they can lead to different but often complementary clustering results. Hence, a consensus approach leveraging the merits of both clustering paradigms could result in a more accurate clustering and a more precise cell type annotation. Results We present scConsensus, an $${\mathbf {R}}$$ R framework for generating a consensus clustering by (1) integrating results from both unsupervised and supervised approaches and (2) refining the consensus clusters using differentially expressed genes. The value of our approach is demonstrated on several existing single-cell RNA sequencing datasets, including data from sorted PBMC sub-populations. Conclusions scConsensus combines the merits of unsupervised and supervised approaches to partition cells with better cluster separation and homogeneity, thereby increasing our confidence in detecting distinct cell types. scConsensus is implemented in $${\mathbf {R}}$$ R and is freely available on GitHub at https://github.com/prabhakarlab/scConsensus.


2020 ◽  
Author(s):  
Yun Zhang ◽  
Brian D. Aevermann ◽  
Trygve E. Bakken ◽  
Jeremy A. Miller ◽  
Rebecca D. Hodge ◽  
...  

AbstractSingle cell/nucleus RNA sequencing (scRNAseq) is emerging as an essential tool to unravel the phenotypic heterogeneity of cells in complex biological systems. While computational methods for scRNAseq cell type clustering have advanced, the ability to integrate datasets to identify common and novel cell types across experiments remains a challenge. Here, we introduce a cluster-to-cluster cell type matching method – FR-Match – that utilizes supervised feature selection for dimensionality reduction and incorporates shared information among cells to determine whether two cell type clusters share the same underlying multivariate gene expression distribution. FR-Match is benchmarked with existing cell-to-cell and cell-to-cluster cell type matching methods using both simulated and real scRNAseq data. FR-Match proved to be a stringent method that produced fewer erroneous matches of distinct cell subtypes and had the unique ability to identify novel cell phenotypes in new datasets. In silico validation demonstrated that the proposed workflow is the only self-contained algorithm that was robust to increasing numbers of true negatives (i.e. non-represented cell types). FR-Match was applied to two human brain scRNAseq datasets sampled from cortical layer 1 and full thickness middle temporal gyrus. When mapping cell types identified in specimens isolated from these overlapping human brain regions, FR-Match precisely recapitulated the laminar characteristics of matched cell type clusters, reflecting their distinct neuroanatomical distributions. An R package and Shiny application are provided at https://github.com/JCVenterInstitute/FRmatch for users to interactively explore and match scRNAseq cell type clusters with complementary visualization tools.


2018 ◽  
Vol 9 (1) ◽  
Author(s):  
Megan Crow ◽  
Anirban Paul ◽  
Sara Ballouz ◽  
Z. Josh Huang ◽  
Jesse Gillis

Sign in / Sign up

Export Citation Format

Share Document