scholarly journals Accurate and fast cell marker gene identification with COSG

2021 ◽  
Author(s):  
Min Dai ◽  
Xiaobing Pei ◽  
Xiu-Jie Wang

Accurate cell classification is the groundwork for downstream analysis of single-cell sequencing data, yet how to identify marker genes to distinguish different cell types still remains as a big challenge. We developed COSG as a cosine similarity-based method for more accurate and scalable marker gene identification. COSG is applicable to single-cell RNA sequencing data, single-cell ATAC sequencing data and spatially resolved transcriptome data. COSG is fast and scalable for ultra-large datasets of million-scale cells. Application on both simulated and real experimental datasets demonstrates the superior performance of COSG in terms of both accuracy and efficiency as compared with other available methods. Marker genes or genomic regions identified by COSG are more indicative and with greater cell-type specificity.

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.


2021 ◽  
Author(s):  
Michael E Nelson ◽  
Simone G Riva ◽  
Ann Cvejic

Spatial transcriptomics is revolutionising the study of single-cell RNA and tissue-wide cell heterogeneity, but few robust methods connecting spatially resolved cells to so-called marker genes from single-cell RNA sequencing, which generate significant insight gleaned from spatial methods, exist. Here we present SMaSH, a general computational framework for extracting key marker genes from single-cell RNA sequencing data for spatial transcriptomics approaches. SMaSH extracts robust and biologically well-motivated marker genes, which characterise the given data-set better than existing and limited computational approaches for global marker gene calculation.


2021 ◽  
Author(s):  
Wenchuan Wang ◽  
Fan Yang ◽  
Yuan Fang ◽  
Duyu Tang ◽  
Junzhou Huang ◽  
...  

AbstractReliable cell type annotation is a prerequisite for downstream analysis of single-cell RNA sequencing data. Existing annotation algorithms typically suffer from improper handling of batch effect, lack of curated marker gene lists, or difficulty in leveraging the latent gene-gene interaction information. Inspired by large scale pretrained langurage models, we present a pretrained deep neural network-based model scBERT (single-cell Bidirectional Encoder Representations from Transformers) to overcome the above challenges. scBERT follows the state-of-the-art paradigm of pre-train and fine-tune in the deep learning field. In the first phase of scBERT, it obtains a general understanding of gene-gene interaction by being pre-trained on huge amounts of unlabeled scRNA-seq data. The pre-trained scBERT can then be used for the cell annotation task of unseen and user-specific scRNA-seq data through supervised fine-tuning. Extensive and rigorous benchmark studies validate the superior performance of scBERT on various tasks, including cell type annotation, novel cell type discovery, as well as investigation of gene-gene interactions. Thus, scBERT enjoys the advantages of improved generalization and interpretability than existing annotation tools.


2021 ◽  
Vol 7 (10) ◽  
pp. eabc5464
Author(s):  
Kiya W. Govek ◽  
Emma C. Troisi ◽  
Zhen Miao ◽  
Rachael G. Aubin ◽  
Steven Woodhouse ◽  
...  

Highly multiplexed immunohistochemistry (mIHC) enables the staining and quantification of dozens of antigens in a tissue section with single-cell resolution. However, annotating cell populations that differ little in the profiled antigens or for which the antibody panel does not include specific markers is challenging. To overcome this obstacle, we have developed an approach for enriching mIHC images with single-cell RNA sequencing data, building upon recent experimental procedures for augmenting single-cell transcriptomes with concurrent antigen measurements. Spatially-resolved Transcriptomics via Epitope Anchoring (STvEA) performs transcriptome-guided annotation of highly multiplexed cytometry datasets. It increases the level of detail in histological analyses by enabling the systematic annotation of nuanced cell populations, spatial patterns of transcription, and interactions between cell types. We demonstrate the utility of STvEA by uncovering the architecture of poorly characterized cell types in the murine spleen using published cytometry and mIHC data of this organ.


2021 ◽  
Author(s):  
Risa Karakida Kawaguchi ◽  
Ziqi Tang ◽  
Stephan Fischer ◽  
Rohit Tripathy ◽  
Peter K. Koo ◽  
...  

Background: Single-cell Assay for Transposase Accessible Chromatin using sequencing (scATAC-seq) measures genome-wide chromatin accessibility for the discovery of cell-type specific regulatory networks. ScATAC-seq combined with single-cell RNA sequencing (scRNA-seq) offers important avenues for ongoing research, such as novel cell-type specific activation of enhancer and transcription factor binding sites as well as chromatin changes specific to cell states. On the other hand, scATAC-seq data is known to be challenging to interpret due to its high number of zeros as well as the heterogeneity derived from different protocols. Because of the stochastic lack of marker gene activities, cell type identification by scATAC-seq remains difficult even at a cluster level. Results: In this study, we exploit reference knowledge obtained from external scATAC-seq or scRNA-seq datasets to define existing cell types and uncover the genomic regions which drive cell-type specific gene regulation. To investigate the robustness of existing cell-typing methods, we collected 7 scATAC-seq datasets targeting mouse brain for a meta-analytic comparison of neuronal cell-type annotation, including a reference atlas generated by the BRAIN Initiative Cell Census Network (BICCN). By comparing the area under the receiver operating characteristics curves (AUROCs) for the three major cell types (inhibitory, excitatory, and non-neuronal cells), cell-typing performance by single markers is found to be highly variable even for known marker genes due to study-specific biases. However, the signal aggregation of a large and redundant marker gene set, optimized via multiple scRNA-seq data, achieves the highest cell-typing performances among 5 existing marker gene sets, from the individual cell to cluster level. That gene set also shows a high consistency with the cluster-specific genes from inhibitory subtypes in two well-annotated datasets, suggesting applicability to rare cell types. Next, we demonstrate a comprehensive assessment of scATAC-seq cell typing using exhaustive combinations of the marker gene sets with supervised learning methods including machine learning classifiers and joint clustering methods. Our results show that the combinations using robust marker gene sets systematically ranked at the top, not only with model based prediction using a large reference data but also with a simple summation of expression strengths across markers. To demonstrate the utility of this robust cell typing approach, we trained a deep neural network to predict chromatin accessibility in each subtype using only DNA sequence. Through model interpretation methods, we identify key motifs enriched about robust gene sets for each neuronal subtype. Conclusions: Through the meta-analytic evaluation of scATAC-seq cell-typing methods, we develop a novel method set to exploit the BICCN reference atlas. Our study strongly supports the value of robust marker gene selection as a feature selection tool and cross-dataset comparison between scATAC-seq datasets to improve alignment of scATAC-seq to known biology. With this novel, high quality epigenetic data, genomic analysis of regulatory regions can reveal sequence motifs that drive cell type-specific regulatory programs.


2020 ◽  
Author(s):  
Edwin Vans ◽  
Ashwini Patil ◽  
Alok Sharma

ABSTRACTAdvances in next-generation sequencing (NGS) have made it possible to carry out transcriptomic studies at single-cell resolution and generate vast amounts of single-cell RNA-seq data rapidly. Thus, tools to analyze this data need to evolve as well to improve accuracy and efficiency. We present FEATS, a python software package that performs clustering on single-cell RNA-seq data. FEATS is capable of performing multiple tasks such as estimating the number of clusters, conducting outlier detection, and integrating data from various experiments. We develop a univariate feature selection based approach for clustering, which involves the selection of top informative features to improve clustering performance. This is motivated by the fact that cell types are often manually determined using the expression of only a few known marker genes. On a variety of single-cell RNA-seq datasets, FEATS gives superior performance compared to the current tools, in terms of adjusted rand index (ARI) and estimating the number of clusters. In addition to cluster estimation, FEATS also performs outlier detection and data integration while giving an excellent computational performance. Thus, FEATS is a comprehensive clustering tool capable of addressing the challenges during the clustering of single-cell RNA-seq data. The installation instructions and documentation of FEATS is available at https://edwinv87.github.io/feats/.


2016 ◽  
Author(s):  
Kieran R Campbell ◽  
Christopher Yau

AbstractPseudotime estimation from single-cell gene expression allows the recovery of temporal information from otherwise static profiles of individual cells. This pseudotemporal information can be used to characterise transient events in temporally evolving biological systems. Conventional algorithms typically emphasise an unsupervised transcriptome-wide approach and use retrospective analysis to evaluate the behaviour of individual genes. Here we introduce an orthogonal approach termed “Ouija” that learns pseudotimes from a small set of marker genes that might ordinarily be used to retrospectively confirm the accuracy of unsupervised pseudotime algorithms. Crucially, we model these genes in terms of switch-like or transient behaviour along the trajectory, allowing us to understand why the pseudotimes have been inferred and learn informative parameters about the behaviour of each gene. Since each gene is associated with a switch or peak time the genes are effectively ordered along with the cells, allowing each part of the trajectory to be understood in terms of the behaviour of certain genes. In the following we introduce our model and demonstrate that in many instances a small panel of marker genes can recover pseudotimes that are consistent with those obtained using the entire transcriptome. Furthermore, we show that our method can detect differences in the regulation timings between two genes and identify “metastable” states - discrete cell types along the continuous trajectories - that recapitulate known cell types. Ouija therefore provides a powerful complimentary approach to existing whole transcriptome based pseudotime estimation methods. An open source implementation is available at http://www.github.com/kieranrcampbell/ouija as an R package and at http://www.github.com/kieranrcampbell/ouijaflow as a Python/TensorFlow package.


2021 ◽  
Author(s):  
Ayshwarya Subramanian ◽  
Mikhail Alperovich ◽  
Bo Li ◽  
Yiming Yang

Quality control (QC) of cells, a critical step in single-cell RNA sequencing data analysis, has largely relied on arbitrarily fixed data-agnostic thresholds on QC metrics such as gene complexity and fraction of reads mapping to mitochondrial genes. The few existing data-driven approaches perform QC at the level of samples or studies without accounting for biological variation in the commonly used QC criteria. We demonstrate that the QC metrics vary both at the tissue and cell state level across technologies, study conditions, and species. We propose data-driven QC (ddqc), an unsupervised adaptive quality control framework that performs flexible and data-driven quality control at the level of cell states while retaining critical biological insights and improved power for downstream analysis. On applying ddqc to 6,228,212 cells and 835 mouse and human samples, we retain a median of 39.7% more cells when compared to conventional data-agnostic QC filters. With ddqc, we recover biologically meaningful trends in gene complexity and ribosomal expression among cell-types enabling exploration of cell states with minimal transcriptional diversity or maximum ribosomal protein expression. Moreover, ddqc allows us to retain cell-types often lost by conventional QC such as metabolically active parenchymal cells, and specialized cells such as neutrophils or gastric chief cells. Taken together, our work proposes a revised paradigm to quality filtering best practices - iterative QC, providing a data-driven quality control framework compatible with observed biological diversity.


2021 ◽  
Author(s):  
Fangfang Yan ◽  
Zhongming Zhao ◽  
Lukas M. Simon

ABSTRACTDroplet-based single-cell RNA sequencing (scRNA-seq) has significantly increased the number of cells profiled per experiment and revolutionized the study of individual transcriptomes. However, to maximize the biological signal robust computational methods are needed to distinguish cell-free from cell-containing droplets. Here, we introduce a novel cell-calling algorithm called EmptyNN, which trains a neural network based on positive-unlabeled learning for improved filtering of barcodes. We leveraged cell hashing and genetic variation to provide ground-truth. EmptyNN accurately removed cell-free droplets while recovering lost cell clusters, and achieved an Area Under the Receiver Operating Characteristics (AUROC) of 94.73% and 96.30%, respectively. The comparisons to current state-of-the-art cell-calling algorithms demonstrated the superior performance of EmptyNN, as measured by the number of recovered cell-containing droplets and cell types. EmptyNN was further applied to two additional datasets and showed good performance. Therefore, EmptyNN represents a powerful tool to enhance scRNA-seq quality control analyses.


2019 ◽  
Author(s):  
Casey A. Thornton ◽  
Ryan M. Mulqueen ◽  
Andrew Nishida ◽  
Kristof A. Torkenczy ◽  
Eve G. Lowenstein ◽  
...  

AbstractHigh-throughput single-cell epigenomic assays can resolve the heterogeneity of cell types and states in complex tissues, however, spatial orientation within the network of interconnected cells is lost. Here, we present a novel method for highly scalable, spatially resolved, single-cell profiling of chromatin states. We use high-density multiregional sampling to perform single-cell combinatorial indexing on Microbiopsies Assigned to Positions for the Assay for Transposase Accessible Chromatin (sciMAP-ATAC) to produce single-cell data of an equivalent quality to non-spatially resolved single-cell ATAC-seq, where each cell is localized to a three-dimensional position within the tissue. A typical experiment comprises between 96 and 384 spatially mapped tissue positions, each producing 10s to over 100 individual single-cell ATAC-seq profiles, and a typical resolution of 214 cubic microns; with the ability to tune the resolution and cell throughput to suit each target application. We apply sciMAP-ATAC to the adult mouse primary somatosensory cortex, where we profile cortical lamination and demonstrate the ability to analyze data from a single tissue position or compare a single cell type in adjacent positions. We also profile the human primary visual cortex, where we produce spatial trajectories through the cortex. Finally, we characterize the spatially progressive nature of cerebral ischemic infarct in the mouse brain using a model of transient middle cerebral artery occlusion. We leverage the spatial information to identify novel and known transcription factor activities that vary by proximity to the ischemic infarction core with cell type specificity.


Sign in / Sign up

Export Citation Format

Share Document