scholarly journals RNA-Sieve: A likelihood-based deconvolution of bulk gene expression data using single-cell references

2020 ◽  
Author(s):  
Dan D. Erdmann-Pham ◽  
Jonathan Fischer ◽  
Justin Hong ◽  
Yun S. Song

AbstractDirect comparison of bulk gene expression profiles is complicated by distinct cell type mixtures in each sample which obscure whether observed differences are actually due to changes in expression levels themselves or simply cell type compositions. Single-cell technology has made it possible to measure gene expression in individual cells, achieving higher resolution at the expense of increased noise. If carefully incorporated, such single-cell data can be used to deconvolve bulk samples to yield accurate estimates of the true cell type proportions, thus enabling one to disentangle the effects of differential expression and cell type mixtures. Here, we propose a generative model and a likelihood-based inference method that uses asymptotic statistical theory and a novel optimization procedure to perform deconvolution of bulk RNA-seq data to produce accurate cell type proportion estimates. We demonstrate the effectiveness of our method, called RNA-Sieve, across a diverse array of scenarios involving real data and discuss several extensions made uniquely possible by our probabilistic framework, including general hypotheses tests and confidence intervals.

Author(s):  
Xiangtao Li ◽  
Shaochuan Li ◽  
Lei Huang ◽  
Shixiong Zhang ◽  
Ka-chun Wong

Abstract Single-cell RNA sequencing (scRNA-seq) technologies have been heavily developed to probe gene expression profiles at single-cell resolution. Deep imputation methods have been proposed to address the related computational challenges (e.g. the gene sparsity in single-cell data). In particular, the neural architectures of those deep imputation models have been proven to be critical for performance. However, deep imputation architectures are difficult to design and tune for those without rich knowledge of deep neural networks and scRNA-seq. Therefore, Surrogate-assisted Evolutionary Deep Imputation Model (SEDIM) is proposed to automatically design the architectures of deep neural networks for imputing gene expression levels in scRNA-seq data without any manual tuning. Moreover, the proposed SEDIM constructs an offline surrogate model, which can accelerate the computational efficiency of the architectural search. Comprehensive studies show that SEDIM significantly improves the imputation and clustering performance compared with other benchmark methods. In addition, we also extensively explore the performance of SEDIM in other contexts and platforms including mass cytometry and metabolic profiling in a comprehensive manner. Marker gene detection, gene ontology enrichment and pathological analysis are conducted to provide novel insights into cell-type identification and the underlying mechanisms. The source code is available at https://github.com/li-shaochuan/SEDIM.


2018 ◽  
Author(s):  
Lingxue Zhu ◽  
Jing Lei ◽  
Bernie Devlin ◽  
Kathryn Roeder

AbstractMotivated by the dynamics of development, in which cells of recognizable types, or pure cell types, transition into other types over time, we propose a method of semi-soft clustering that can classify both pure and intermediate cell types from data on gene expression or protein abundance from individual cells. Called SOUP, for Semi-sOft clUstering with Pure cells, this novel algorithm reveals the clustering structure for both pure cells, which belong to one single cluster, as well as transitional cells with soft memberships. SOUP involves a two-step process: identify the set of pure cells and then estimate a membership matrix. To find pure cells, SOUP uses the special block structure the K cell types form in a similarity matrix, devised by pairwise comparison of the gene expression profiles of individual cells. Once pure cells are identified, they provide the key information from which the membership matrix can be computed. SOUP is applicable to general clustering problems as well, as long as the unrestrictive modeling assumptions hold. The performance of SOUP is documented via extensive simulation studies. Using SOUP to analyze two single cell data sets from brain shows it produce sensible and interpretable results.


Author(s):  
Meichen Dong ◽  
Aatish Thennavan ◽  
Eugene Urrutia ◽  
Yun Li ◽  
Charles M Perou ◽  
...  

Abstract Recent advances in single-cell RNA sequencing (scRNA-seq) enable characterization of transcriptomic profiles with single-cell resolution and circumvent averaging artifacts associated with traditional bulk RNA sequencing (RNA-seq) data. Here, we propose SCDC, a deconvolution method for bulk RNA-seq that leverages cell-type specific gene expression profiles from multiple scRNA-seq reference datasets. SCDC adopts an ENSEMBLE method to integrate deconvolution results from different scRNA-seq datasets that are produced in different laboratories and at different times, implicitly addressing the problem of batch-effect confounding. SCDC is benchmarked against existing methods using both in silico generated pseudo-bulk samples and experimentally mixed cell lines, whose known cell-type compositions serve as ground truths. We show that SCDC outperforms existing methods with improved accuracy of cell-type decomposition under both settings. To illustrate how the ENSEMBLE framework performs in complex tissues under different scenarios, we further apply our method to a human pancreatic islet dataset and a mouse mammary gland dataset. SCDC returns results that are more consistent with experimental designs and that reproduce more significant associations between cell-type proportions and measured phenotypes.


2019 ◽  
Author(s):  
Meichen Dong ◽  
Aatish Thennavan ◽  
Eugene Urrutia ◽  
Yun Li ◽  
Charles M. Perou ◽  
...  

AbstractRecent advances in single-cell RNA sequencing (scRNA-seq) enable characterization of transcriptomic profiles with single-cell resolution and circumvent averaging artifacts associated with traditional bulk RNA sequencing (RNA-seq) data. Here, we propose SCDC, a deconvolution method for bulk RNA-seq that leverages cell-type specific gene expression profiles from multiple scRNA-seq reference datasets. SCDC adopts an ENSEMBLE method to integrate deconvolution results from different scRNA-seq datasets that are produced in different laboratories and at different times, implicitly addressing the problem of batch-effect confounding. SCDC is benchmarked against existing methods using both in silico generated pseudo-bulk samples and experimentally mixed cell lines, whose known cell-type compositions serve as ground truths. We show that SCDC outperforms existing methods with improved accuracy of cell-type decomposition under both settings. To illustrate how the ENSEMBLE framework performs in complex tissues under different scenarios, we further apply our method to a human pancreatic islet dataset and a mouse mammary gland dataset. SCDC returns results that are more consistent with experimental designs and that reproduce more significant associations between cell-type proportions and measured phenotypes.


Author(s):  
Isabella N. Grabski ◽  
Rafael A. Irizarry

AbstractSingle-cell RNA sequencing (scRNA-seq) quantifies gene expression for individual cells in a sample, which allows distinct cell-type populations to be identified and characterized. An important step in many scRNA-seq analysis pipelines is the annotation of cells into known cell-types. While this can be achieved using experimental techniques, such as fluorescence-activated cell sorting, these approaches are impractical for large numbers of cells. This motivates the development of data-driven cell-type annotation methods. We find limitations with current approaches due to the reliance on known marker genes or from overfitting because of systematic differences between studies or batch effects. Here, we present a statistical approach that leverages public datasets to combine information across thousands of genes, uses a latent variable model to define cell-type-specific barcodes and account for batch effect variation, and probabilistically annotates cell-type identity. The barcoding approach also provides a new way to discover marker genes. Using a range of datasets, including those generated to represent imperfect real-world reference data, we demonstrate that our approach substantially outperforms current reference-based methods, in particular when predicting across studies. Our approach also demonstrates that current approaches based on unsupervised clustering lead to false discoveries related to novel cell-types.


2020 ◽  
Vol 49 (D1) ◽  
pp. D1413-D1419 ◽  
Author(s):  
Tianyi Zhao ◽  
Shuxuan Lyu ◽  
Guilin Lu ◽  
Liran Juan ◽  
Xi Zeng ◽  
...  

Abstract SC2disease (http://easybioai.com/sc2disease/) is a manually curated database that aims to provide a comprehensive and accurate resource of gene expression profiles in various cell types for different diseases. With the development of single-cell RNA sequencing (scRNA-seq) technologies, uncovering cellular heterogeneity of different tissues for different diseases has become feasible by profiling transcriptomes across cell types at the cellular level. In particular, comparing gene expression profiles between different cell types and identifying cell-type-specific genes in various diseases offers new possibilities to address biological and medical questions. However, systematic, hierarchical and vast databases of gene expression profiles in human diseases at the cellular level are lacking. Thus, we reviewed the literature prior to March 2020 for studies which used scRNA-seq to study diseases with human samples, and developed the SC2disease database to summarize all the data by different diseases, tissues and cell types. SC2disease documents 946 481 entries, corresponding to 341 cell types, 29 tissues and 25 diseases. Each entry in the SC2disease database contains comparisons of differentially expressed genes between different cell types, tissues and disease-related health status. Furthermore, we reanalyzed gene expression matrix by unified pipeline to improve the comparability between different studies. For each disease, we also compare cell-type-specific genes with the corresponding genes of lead single nucleotide polymorphisms (SNPs) identified in genome-wide association studies (GWAS) to implicate cell type specificity of the traits.


Author(s):  
Johan Gustafsson ◽  
Felix Held ◽  
Jonathan Robinson ◽  
Elias Björnson ◽  
Rebecka Jörnsten ◽  
...  

Abstract Background Cell-type specific gene expression profiles are needed for many computational methods operating on bulk RNA-Seq samples, such as deconvolution of cell-type fractions and digital cytometry. However, the gene expression profile of a cell type can vary substantially due to both technical factors and biological differences in cell state and surroundings, reducing the efficacy of such methods. Here, we investigated which factors contribute most to this variation. Results We evaluated different normalization methods, quantified the magnitude of variation introduced by different sources, and examined the differences between UMI-based single-cell RNA-Seq and bulk RNA-Seq. We applied methods such as random forest regression to a collection of publicly available bulk and single-cell RNA-Seq datasets containing B and T cells, and found that the technical variation across laboratories is of the same magnitude as the biological variation across cell types. Tissue of origin and cell subtype are less important but still substantial factors, while the difference between individuals is relatively small. We also show that much of the differences between UMI-based single-cell and bulk RNA-Seq methods can be explained by the number of read duplicates per mRNA molecule in the single-cell sample.Conclusions Our work shows the importance of either matching or correcting for technical factors when creating cell-type specific gene expression profiles that are to be used together with bulk samples.


2020 ◽  
Author(s):  
Sooyoun Oh ◽  
Haesun Park ◽  
Xiuwei Zhang

AbstractRecent advances in single cell transcriptomics have allowed us to examine the identify of each single cell, thus have led to discovery of new cell types and provide a high resolution map of cell type composition in tissues. Technologies which can measure another type of data of a single cell in addition to the gene-expression data provide a more comprehensive picture of a cell, and meanwhile pose challenges for data integration tasks. We consider the spatial location of cells, which is an important feature of cells, combined with the cells’ gene-expression profiles, to determine the cell type identity. We aim to jointly classify cells based on their locations relative to other cells in the system as well as their gene expression profiles. We have developed scHybridNMF (single-cell Hybrid Nonnegative Matrix Factorization), which performs cell type identification by incorporating single cell gene expression data with cell location data. We combined two classical methods, nonnegative matrix factorization with a k-means clustering scheme, to respectively represent high-dimensional gene expression data and low-dimensional location data together. Our method incorporates a novel cell location term to the gene expression clustering. We show that scHybridNMF can make use of the location data to improve cell type clustering. In particular, we show that under multiple scenarios, including that when the number of genes profiled is low, and when the location data is noisy, scHybridNMF outperforms the standalone algorithms NMF and k-means, and an existing method HMRF which also uses cell location and gene-expression data for cell type identification.


2020 ◽  
Author(s):  
Johan Gustafsson ◽  
Felix Held ◽  
Jonathan Robinson ◽  
Elias Björnson ◽  
Rebecka Jörnsten ◽  
...  

Abstract Cell-type specific gene expression profiles are needed for many computational methods operating on bulk RNA-Seq samples, such as deconvolution of cell-type fractions and digital cytometry. However, the gene expression profile of a cell type can vary substantially due to both technical factors and biological differences in cell state and surroundings, reducing the efficacy of such methods. Here, we investigated which factors contribute most to this variation. We evaluated different normalization methods, quantified the variance explained by different factors, evaluated the effect on deconvolution of cell type fractions, and examined the differences between UMI-based single-cell RNA-Seq and bulk RNA-Seq. We investigated a collection of publicly available bulk and single-cell RNA-Seq datasets containing B and T cells, and found that the technical variation across laboratories is substantial, even for genes specifically selected for deconvolution, and has a confounding effect on deconvolution. Tissue of origin is also a substantial factor, highlighting the challenge of applying cell type profiles derived from blood on mixtures from other tissues. We also show that much of the differences between UMI-based single-cell and bulk RNA-Seq methods can be explained by the number of read duplicates per mRNA molecule in the single-cell sample. Our work shows the importance of either matching or correcting for technical factors when creating cell-type specific gene expression profiles that are to be used together with bulk samples.


Sign in / Sign up

Export Citation Format

Share Document