scholarly journals Optimal marker gene selection for cell type discrimination in single cell analyses

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Bianca Dumitrascu ◽  
Soledad Villar ◽  
Dustin G. Mixon ◽  
Barbara E. Engelhardt

AbstractSingle-cell technologies characterize complex cell populations across multiple data modalities at unprecedented scale and resolution. Multi-omic data for single cell gene expression, in situ hybridization, or single cell chromatin states are increasingly available across diverse tissue types. When isolating specific cell types from a sample of disassociated cells or performing in situ sequencing in collections of heterogeneous cells, one challenging task is to select a small set of informative markers that robustly enable the identification and discrimination of specific cell types or cell states as precisely as possible. Given single cell RNA-seq data and a set of cellular labels to discriminate, scGeneFit selects gene markers that jointly optimize cell label recovery using label-aware compressive classification methods. This results in a substantially more robust and less redundant set of markers than existing methods, most of which identify markers that separate each cell label from the rest. When applied to a data set given a hierarchy of cell types as labels, the markers found by our method improves the recovery of the cell type hierarchy with fewer markers than existing methods using a computationally efficient and principled optimization.

2019 ◽  
Author(s):  
Bianca Dumitrascu ◽  
Soledad Villar ◽  
Dustin G. Mixon ◽  
Barbara E. Engelhardt

Single-cell technologies characterize complex cell populations across multiple data modalities at un-precedented scale and resolution. Multi-omic data for single cell gene expression,in situhybridization, or single cell chromatin states are increasingly available across diverse tissue types. When isolating specific cell types from a sample of disassociated cells or performingin situsequencing in collections of heterogeneous cells, one challenging task is to select a small set of informative markers to identify and differentiate specific cell types or cell states as precisely as possible. Given single cell RNA-seq data and a set of cellular labels to discriminate, scGene-Fit selects gene transcript markers that jointly optimize cell label recovery using label-aware compressive classification methods, resulting in a substantially more robust and less redundant set of markers than existing methods. When applied to a data set given a hierarchy of cell type labels, the markers found by our method enable the recovery of the label hierarchy through a computationally efficient and principled optimization.


BMC Biology ◽  
2020 ◽  
Vol 18 (1) ◽  
Author(s):  
Elin Lundin ◽  
Chenglin Wu ◽  
Albin Widmark ◽  
Mikaela Behm ◽  
Jens Hjerling-Leffler ◽  
...  

Abstract Background Adenosine-to-inosine (A-to-I) RNA editing is a process that contributes to the diversification of proteins that has been shown to be essential for neurotransmission and other neuronal functions. However, the spatiotemporal and diversification properties of RNA editing in the brain are largely unknown. Here, we applied in situ sequencing to distinguish between edited and unedited transcripts in distinct regions of the mouse brain at four developmental stages, and investigate the diversity of the RNA landscape. Results We analyzed RNA editing at codon-altering sites using in situ sequencing at single-cell resolution, in combination with the detection of individual ADAR enzymes and specific cell type marker transcripts. This approach revealed cell-type-specific regulation of RNA editing of a set of transcripts, and developmental and regional variation in editing levels for many of the targeted sites. We found increasing editing diversity throughout development, which arises through regional- and cell type-specific regulation of ADAR enzymes and target transcripts. Conclusions Our single-cell in situ sequencing method has proved useful to study the complex landscape of RNA editing and our results indicate that this complexity arises due to distinct mechanisms of regulating individual RNA editing sites, acting both regionally and in specific cell types.


2018 ◽  
Author(s):  
Brian Hie ◽  
Bryan Bryson ◽  
Bonnie Berger

AbstractResearchers are generating single-cell RNA sequencing (scRNA-seq) profiles of diverse biological systems1–4 and every cell type in the human body.5 Leveraging this data to gain unprecedented insight into biology and disease will require assembling heterogeneous cell populations across multiple experiments, laboratories, and technologies. Although methods for scRNA-seq data integration exist6,7, they often naively merge data sets together even when the data sets have no cell types in common, leading to results that do not correspond to real biological patterns. Here we present Scanorama, inspired by algorithms for panorama stitching, that overcomes the limitations of existing methods to enable accurate, heterogeneous scRNA-seq data set integration. Our strategy identifies and merges the shared cell types among all pairs of data sets and is orders of magnitude faster than existing techniques. We use Scanorama to combine 105,476 cells from 26 diverse scRNA-seq experiments across 9 different technologies into a single comprehensive reference, demonstrating how Scanorama can be used to obtain a more complete picture of cellular function across a wide range of scRNA-seq experiments.


2018 ◽  
Author(s):  
Wennan Chang ◽  
Changlin Wan ◽  
Xiaoyu Lu ◽  
Szu-wei Tu ◽  
Yifan Sun ◽  
...  

AbstractWe developed a novel deconvolution method, namely Inference of Cell Types and Deconvolution (ICTD) that addresses the fundamental issue of identifiability and robustness in current tissue data deconvolution problem. ICTD provides substantially new capabilities for omics data based characterization of a tissue microenvironment, including (1) maximizing the resolution in identifying resident cell and sub types that truly exists in a tissue, (2) identifying the most reliable marker genes for each cell type, which are tissue and data set specific, (3) handling the stability problem with co-linear cell types, (4) co-deconvoluting with available matched multi-omics data, and (5) inferring functional variations specific to one or several cell types. ICTD is empowered by (i) rigorously derived mathematical conditions of identifiable cell type and cell type specific functions in tissue transcriptomics data and (ii) a semi supervised approach to maximize the knowledge transfer of cell type and functional marker genes identified in single cell or bulk cell data in the analysis of tissue data, and (iii) a novel unsupervised approach to minimize the bias brought by training data. Application of ICTD on real and single cell simulated tissue data validated that the method has consistently good performance for tissue data coming from different species, tissue microenvironments, and experimental platforms. Other than the new capabilities, ICTD outperformed other state-of-the-art devolution methods on prediction accuracy, the resolution of identifiable cell, detection of unknown sub cell types, and assessment of cell type specific functions. The premise of ICTD also lies in characterizing cell-cell interactions and discovering cell types and prognostic markers that are predictive of clinical outcomes.


Author(s):  
Xiaoyu Lu ◽  
Szu-Wei Tu ◽  
Wennan Chang ◽  
Changlin Wan ◽  
Jiashi Wang ◽  
...  

Abstract Deconvolution of mouse transcriptomic data is challenged by the fact that mouse models carry various genetic and physiological perturbations, making it questionable to assume fixed cell types and cell type marker genes for different data set scenarios. We developed a Semi-Supervised Mouse data Deconvolution (SSMD) method to study the mouse tissue microenvironment. SSMD is featured by (i) a novel nonparametric method to discover data set-specific cell type signature genes; (ii) a community detection approach for fixing cell types and their marker genes; (iii) a constrained matrix decomposition method to solve cell type relative proportions that is robust to diverse experimental platforms. In summary, SSMD addressed several key challenges in the deconvolution of mouse tissue data, including: (i) varied cell types and marker genes caused by highly divergent genotypic and phenotypic conditions of mouse experiment; (ii) diverse experimental platforms of mouse transcriptomics data; (iii) small sample size and limited training data source and (iv) capable to estimate the proportion of 35 cell types in blood, inflammatory, central nervous or hematopoietic systems. In silico and experimental validation of SSMD demonstrated its high sensitivity and accuracy in identifying (sub) cell types and predicting cell proportions comparing with state-of-the-arts methods. A user-friendly R package and a web server of SSMD are released via https://github.com/xiaoyulu95/SSMD.


2019 ◽  
Author(s):  
Arnav Moudgil ◽  
Michael N. Wilkinson ◽  
Xuhua Chen ◽  
June He ◽  
Alex J. Cammack ◽  
...  

AbstractIn situ measurements of transcription factor (TF) binding are confounded by cellular heterogeneity and represent averaged profiles in complex tissues. Single cell RNA-seq (scRNA-seq) is capable of resolving different cell types based on gene expression profiles, but no technology exists to directly link specific cell types to the binding pattern of TFs in those cell types. Here, we present self-reporting transposons (SRTs) and their use in single cell calling cards (scCC), a novel assay for simultaneously capturing gene expression profiles and mapping TF binding sites in single cells. First, we show how the genomic locations of SRTs can be recovered from mRNA. Next, we demonstrate that SRTs deposited by the piggyBac transposase can be used to map the genome-wide localization of the TFs SP1, through a direct fusion of the two proteins, and BRD4, through its native affinity for piggyBac. We then present the scCC method, which maps SRTs from scRNA-seq libraries, thus enabling concomitant identification of cell types and TF binding sites in those same cells. As a proof-of-concept, we show recovery of cell type-specific BRD4 and SP1 binding sites from cultured cells. Finally, we map Brd4 binding sites in the mouse cortex at single cell resolution, thus establishing a new technique for studying TF biology in situ.


2020 ◽  
Author(s):  
Tao Yang ◽  
Nicole Alessandri-Haber ◽  
Wen Fury ◽  
Michael Schaner ◽  
Robert Breese ◽  
...  

AbstractRNA sequencing technology promises an unprecedented opportunity in learning disease mechanisms and discovering new treatment targets. Recent spatial transcriptomics methods further enable the transcriptome profiling at spatially resolved spots in a tissue section. In controlled experiments, it is often of immense importance to know the cell composition in different samples. Understanding the cell type content in each tissue spot is also crucial to the spatial transcriptome data interpretation. Though single cell RNA-seq has the power to reveal cell type composition and expression heterogeneity in different cells, it remains costly and sometimes infeasible when live cells cannot be obtained or sufficiently dissociated. To computationally resolve the cell composition in RNA-seq data of mixed cells, we present AdRoit, an accurate androbust method to infer transcriptome composition. The method estimates the proportions of each cell type in the compound RNA-seq data using known single cell data of relevant cell types. It uniquely uses an adaptive learning approach to correct the bias gene-wise due to the difference in sequencing techniques. AdRoit also utilizes cell type specific genes while control their cross-sample variability. Our systematic benchmarking, spanning from simple to complex tissues, shows that AdRoit has superior sensitivity and specificity compared to other existing methods. Its performance holds for multiple single cell and compound RNA-seq platforms. In addition, AdRoit is computationally efficient and runs one to two orders of magnitude faster than some of the state-of-the-art methods.


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.


2021 ◽  
Author(s):  
Carolyn Shasha ◽  
Yuan Tian ◽  
Florian Mair ◽  
Helen E Rodgers Miller ◽  
Raphael Gottardo

Automated cell type annotation of single-cell RNA-seq data has the potential to significantly improve and streamline single cell data analysis, facilitating comparisons and meta-analyses. However, many of the current state-of-the-art techniques suffer from limitations, such as reliance on a single reference dataset or marker gene set, or excessive run times for large datasets. Acquiring high-quality labeled data to use as a reference can be challenging. With CITE-seq, surface protein expression of cells can be directly measured in addition to the RNA expression, facilitating cell type annotation. Here, we compiled and annotated a collection of 16 publicly available CITE-seq datasets. This data was then used as training data to develop Superscan, a supervised machine learning-based prediction model. Using our 16 reference datasets, we benchmarked Superscan and showed that it performs better in terms of both accuracy and speed when compared to other state-of-the-art cell annotation methods. Superscan is pre-trained on a collection of primarily PBMC immune datasets; however, additional data and cell types can be easily added to the training data for further improvement. Finally, we used Superscan to reanalyze a previously published dataset, demonstrating its applicability even when the dataset includes cell types that are missing from the training set.


2020 ◽  
Author(s):  
Ajay Patil ◽  
Ashwini Patil

AbstractSingle-cell RNA-seq is widely used to study transcriptional patterns of genes in individual cells. In spite of current advances in technology, assigning cell types in single-cell datasets remains a bottleneck due to the lack of a comprehensive reference database and a fast search method in a single tool. CellKb Immune is a knowledgebase of manually collected, curated and annotated marker gene sets from cell types in the mammalian immune response. It finds matching cell types in literature given a list of genes using a novel rank-based algorithm optimized for rapid searching across marker gene lists of differing lengths. We evaluated the contents and search algorithm of CellKb Immune using a leave-one-out approach. We further used CellKb Immune to annotate previously defined marker gene sets from Immgen to confirm its accuracy and coverage. CellKb Immune provides an easy to use database with a fast and reliable method to find matching cell types and annotate cells in single-cell experiments in a single tool. It is available at https://www.cellkb.com/immune.


Sign in / Sign up

Export Citation Format

Share Document