scholarly journals bigSCale: An Analytical Framework for Big-Scale Single-Cell Data

2017 ◽  
Author(s):  
Giovanni Iacono ◽  
Elisabetta Mereu ◽  
Amy Guillaumet-Adkins ◽  
Roser Corominas ◽  
Ivon Cuscó ◽  
...  

AbstractSingle-cell RNA sequencing significantly deepened our insights into complex tissues and latest techniques are capable processing ten-thousands of cells simultaneously. With bigSCale, we provide an analytical framework being scalable to analyze millions of cells, addressing challenges of future large datasets. Unlike previous methods, bigSCale does not constrain data to fit an a priori-defined distribution and instead uses an accurate numerical model of noise. We evaluated the performance of bigSCale using a biological model of aberrant gene expression in patient derived neuronal progenitor cells and simulated datasets, which underlined its speed and accuracy in differential expression analysis. We further applied bigSCale to analyze 1.3 million cells from the mouse developing forebrain. Herein, we identified rare populations, such as Reelin positive Cajal-Retzius neurons, for which we determined a previously not recognized heterogeneity associated to distinct differentiation stages, spatial organization and cellular function. Together, bigSCale presents a perfect solution to address future challenges of large single-cell datasets.Extended AbstractSingle-cell RNA sequencing (scRNAseq) significantly deepened our insights into complex tissues by providing high-resolution phenotypes for individual cells. Recent microfluidic-based methods are scalable to ten-thousands of cells, enabling an unbiased sampling and comprehensive characterization without prior knowledge. Increasing cell numbers, however, generates extremely big datasets, which extends processing time and challenges computing resources. Current scRNAseq analysis tools are not designed to analyze datasets larger than from thousands of cells and often lack sensitivity and specificity to identify marker genes for cell populations or experimental conditions. With bigSCale, we provide an analytical framework for the sensitive detection of population markers and differentially expressed genes, being scalable to analyze millions of single cells. Unlike other methods that use simple or mixture probabilistic models with negative binomial, gamma or Poisson distributions to handle the noise and sparsity of scRNAseq data, bigSCale does not constrain the data to fit an a priori-defined distribution. Instead, bigSCale uses large sample sizes to estimate a highly accurate and comprehensive numerical model of noise and gene expression. The framework further includes modules for differential expression (DE) analysis, cell clustering and population marker identification. Moreover, a directed convolution strategy allows processing of extremely large data sets, while preserving the transcript information from individual cells.We evaluate the performance of bigSCale using a biological model for reduced or elevated gene expression levels. Specifically, we perform scRNAseq of 1,920 patient derived neuronal progenitor cells from Williams-Beuren and 7q11.23 microduplication syndrome patients, harboring a deletion or duplication of 7q11.23, respectively. The affected region contains 28 genes whose transcriptional levels vary in line with their allele frequency. BigSCale detects expression changes with respect to cells from a healthy donor and outperforms other methods for single-cell DE analysis in sensitivity. Simulated data sets, underline the performance of bigSCale in DE analysis as it is faster and more sensitive and specific than other methods. The probabilistic model of cell-distances within bigSCale is further suitable for unsupervised clustering and the identification of cell types and subpopulations. Using bigSCale, we identify all major cell types of the somatosensory cortex and hippocampus analyzing 3,005 cells from adult mouse brains. Remarkably, we increase the number of cell population specific marker genes 4-6-fold compared to the original analysis and, moreover, define markers of higher order cell types. These include CD90 (Thy1), a neuronal surface receptor, potentially suitable for isolating intact neurons from complex brain samples.To test its applicability for large data sets, we apply bigSCale on scRNAseq data from 1.3 million cells derived from the pallium of the mouse developing forebrain (E18, 10x Genomics). Our directed down-sampling strategy accumulates transcript counts from cells with similar transcriptional profiles into index cell transcriptomes, thereby defining cellular clusters with improved resolution. Accordingly, index cell clusters provide a rich resource of marker genes for the main brain cell types and less frequent subpopulations. Our analysis of rare populations includes poorly characterized developmental cell types, such as neuron progenitors from the subventricular zone and neocortical Reelin positive neurons known as Cajal-Retzius (CR) cells. The latter represent a transient population which regulates the laminar formation of the developing neocortex and whose malfunctioning causes major neurodevelopmental disorders like autism or schizophrenia. Most importantly, index cell cluster can be deconvoluted to individual cell level for targeted analysis of populations of interest. Through decomposition of Reelin positive neurons, we determined a previously not recognized heterogeneity among CR cells, which we could associate to distinct differentiation stages as well as spatial and functional differences in the developing mouse brain. Specifically, subtypes of CR cells identified by bigSCale express different compositions of NMDA, AMPA and glycine receptor subunits, pointing to subpopulations with distinct membrane properties. Furthermore, we found Cxcl12, a chemokine secreted by the meninges and regulating the tangential migration of CR cells, to be also expressed in CR cells located in the marginal zone of the neocortex, indicating a self-regulated migration capacity.Together, bigSCale presents a perfect solution for the processing and analysis of scRNAseq data from millions of single cells. Its speed and sensitivity makes it suitable to the address future challenges of large single-cell data sets.

2018 ◽  
Author(s):  
Changlin Wan ◽  
Wennan Chang ◽  
Yu Zhang ◽  
Fenil Shah ◽  
Xiaoyu Lu ◽  
...  

ABSTRACTA key challenge in modeling single-cell RNA-seq (scRNA-seq) data is to capture the diverse gene expression states regulated by different transcriptional regulatory inputs across single cells, which is further complicated by a large number of observed zero and low expressions. We developed a left truncated mixture Gaussian (LTMG) model that stems from the kinetic relationships between the transcriptional regulatory inputs and metabolism of mRNA and gene expression abundance in a cell. LTMG infers the expression multi-modalities across single cell entities, representing a gene’s diverse expression states; meanwhile the dropouts and low expressions are treated as left truncated, specifically representing an expression state that is under suppression. We demonstrated that LTMG has significantly better goodness of fitting on an extensive number of single-cell data sets, comparing to three other state of the art models. In addition, our systems kinetic approach of handling the low and zero expressions and correctness of the identified multimodality are validated on several independent experimental data sets. Application on data of complex tissues demonstrated the capability of LTMG in extracting varied expression states specific to cell types or cell functions. Based on LTMG, a differential gene expression test and a co-regulation module identification method, namely LTMG-DGE and LTMG-GCR, are further developed. We experimentally validated that LTMG-DGE is equipped with higher sensitivity and specificity in detecting differentially expressed genes, compared with other five popular methods, and that LTMG-GCR is capable to retrieve the gene co-regulation modules corresponding to perturbed transcriptional regulations. A user-friendly R package with all the analysis power is available at https://github.com/zy26/LTMGSCA.


2019 ◽  
Vol 47 (18) ◽  
pp. e111-e111 ◽  
Author(s):  
Changlin Wan ◽  
Wennan Chang ◽  
Yu Zhang ◽  
Fenil Shah ◽  
Xiaoyu Lu ◽  
...  

Abstract A key challenge in modeling single-cell RNA-seq data is to capture the diversity of gene expression states regulated by different transcriptional regulatory inputs across individual cells, which is further complicated by largely observed zero and low expressions. We developed a left truncated mixture Gaussian (LTMG) model, from the kinetic relationships of the transcriptional regulatory inputs, mRNA metabolism and abundance in single cells. LTMG infers the expression multi-modalities across single cells, meanwhile, the dropouts and low expressions are treated as left truncated. We demonstrated that LTMG has significantly better goodness of fitting on an extensive number of scRNA-seq data, comparing to three other state-of-the-art models. Our biological assumption of the low non-zero expressions, rationality of the multimodality setting, and the capability of LTMG in extracting expression states specific to cell types or functions, are validated on independent experimental data sets. A differential gene expression test and a co-regulation module identification method are further developed. We experimentally validated that our differential expression test has higher sensitivity and specificity, compared with other five popular methods. The co-regulation analysis is capable of retrieving gene co-regulation modules corresponding to perturbed transcriptional regulations. A user-friendly R package with all the analysis power is available at https://github.com/zy26/LTMGSCA.


2018 ◽  
Author(s):  
Christopher Heje Grønbech ◽  
Maximillian Fornitz Vording ◽  
Pascal Timshel ◽  
Casper Kaae Sønderby ◽  
Tune Hannes Pers ◽  
...  

AbstractMotivationModels for analysing and making relevant biological inferences from massive amounts of complex single-cell transcriptomic data typically require several individual data-processing steps, each with their own set of hyperparameter choices. With deep generative models one can work directly with count data, make likelihood-based model comparison, learn a latent representation of the cells and capture more of the variability in different cell populations.ResultsWe propose a novel method based on variational auto-encoders (VAEs) for analysis of single-cell RNA sequencing (scRNA-seq) data. It avoids data preprocessing by using raw count data as input and can robustly estimate the expected gene expression levels and a latent representation for each cell. We tested several count likelihood functions and a variant of the VAE that has a priori clustering in the latent space. We show for several scRNA-seq data sets that our method outperforms recently proposed scRNA-seq methods in clustering cells and that the resulting clusters reflect cell types.Availability and implementationOur method, called scVAE, is implemented in Python using the TensorFlow machine-learning library, and it is freely available at https://github.com/scvae/scvae.


2020 ◽  
Vol 36 (12) ◽  
pp. 3910-3912 ◽  
Author(s):  
Oscar Franzén ◽  
Johan L M Björkegren

Abstract Summary Single-cell RNA sequencing (scRNA-seq) is a technology to measure gene expression in single cells. It has enabled discovery of new cell types and established cell type atlases of tissues and organs. The widespread adoption of scRNA-seq has created a need for user-friendly software for data analysis. We have developed a web server, alona that incorporates several of the most popular single-cell analysis algorithms into a flexible pipeline. alona can perform quality filtering, normalization, batch correction, clustering, cell type annotation and differential gene expression analysis. Data are visualized in the web browser using an interface based on JavaScript, allowing the user to query genes of interest and visualize the cluster structure. alona accepts a compressed gene expression matrix and identifies cell clusters with a graph-based clustering strategy. Cell types are identified from a comprehensive collection of marker genes or by specifying a custom set of marker genes. Availability and implementation The service runs at https://alona.panglaodb.se and the Python package can be downloaded from https://oscar-franzen.github.io/adobo/. Supplementary information Supplementary data are available at Bioinformatics online.


2019 ◽  
Author(s):  
Marcus Alvarez ◽  
Elior Rahmani ◽  
Brandon Jew ◽  
Kristina M. Garske ◽  
Zong Miao ◽  
...  

AbstractSingle-nucleus RNA sequencing (snRNA-seq) measures gene expression in individual nuclei instead of cells, allowing for unbiased cell type characterization in solid tissues. Contrary to single-cell RNA seq (scRNA-seq), we observe that snRNA-seq is commonly subject to contamination by high amounts of extranuclear background RNA, which can lead to identification of spurious cell types in downstream clustering analyses if overlooked. We present a novel approach to remove debris-contaminated droplets in snRNA-seq experiments, called Debris Identification using Expectation Maximization (DIEM). Our likelihood-based approach models the gene expression distribution of debris and cell types, which are estimated using EM. We evaluated DIEM using three snRNA-seq data sets: 1) human differentiating preadipocytes in vitro, 2) fresh mouse brain tissue, and 3) human frozen adipose tissue (AT) from six individuals. All three data sets showed various degrees of extranuclear RNA contamination. We observed that existing methods fail to account for contaminated droplets and led to spurious cell types. When compared to filtering using these state of the art methods, DIEM better removed droplets containing high levels of extranuclear RNA and led to higher quality clusters. Although DIEM was designed for snRNA-seq data, we also successfully applied DIEM to single-cell data. To conclude, our novel method DIEM removes debris-contaminated droplets from single-cell-based data fast and effectively, leading to cleaner downstream analysis. Our code is freely available for use at https://github.com/marcalva/diem.


2018 ◽  
Author(s):  
Douglas Abrams ◽  
Parveen Kumar ◽  
R. Krishna Murthy Karuturi ◽  
Joshy George

AbstractBackgroundThe advent of single cell RNA sequencing (scRNA-seq) enabled researchers to study transcriptomic activity within individual cells and identify inherent cell types in the sample. Although numerous computational tools have been developed to analyze single cell transcriptomes, there are no published studies and analytical packages available to guide experimental design and to devise suitable analysis procedure for cell type identification.ResultsWe have developed an empirical methodology to address this important gap in single cell experimental design and analysis into an easy-to-use tool called SCEED (Single Cell Empirical Experimental Design and analysis). With SCEED, user can choose a variety of combinations of tools for analysis, conduct performance analysis of analytical procedures and choose the best procedure, and estimate sample size (number of cells to be profiled) required for a given analytical procedure at varying levels of cell type rarity and other experimental parameters. Using SCEED, we examined 3 single cell algorithms using 48 simulated single cell datasets that were generated for varying number of cell types and their proportions, number of genes expressed per cell, number of marker genes and their fold change, and number of single cells successfully profiled in the experiment.ConclusionsBased on our study, we found that when marker genes are expressed at fold change of 4 or more than the rest of the genes, either Seurat or Simlr algorithm can be used to analyze single cell dataset for any number of single cells isolated (minimum 1000 single cells were tested). However, when marker genes are expected to be only up to fC 2 upregulated, choice of the single cell algorithm is dependent on the number of single cells isolated and proportion of rare cell type to be identified. In conclusion, our work allows the assessment of various single cell methods and also aids in examining the single cell experimental design.


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.


2018 ◽  
Vol 29 (8) ◽  
pp. 2060-2068 ◽  
Author(s):  
Nikos Karaiskos ◽  
Mahdieh Rahmatollahi ◽  
Anastasiya Boltengagen ◽  
Haiyue Liu ◽  
Martin Hoehne ◽  
...  

Background Three different cell types constitute the glomerular filter: mesangial cells, endothelial cells, and podocytes. However, to what extent cellular heterogeneity exists within healthy glomerular cell populations remains unknown.Methods We used nanodroplet-based highly parallel transcriptional profiling to characterize the cellular content of purified wild-type mouse glomeruli.Results Unsupervised clustering of nearly 13,000 single-cell transcriptomes identified the three known glomerular cell types. We provide a comprehensive online atlas of gene expression in glomerular cells that can be queried and visualized using an interactive and freely available database. Novel marker genes for all glomerular cell types were identified and supported by immunohistochemistry images obtained from the Human Protein Atlas. Subclustering of endothelial cells revealed a subset of endothelium that expressed marker genes related to endothelial proliferation. By comparison, the podocyte population appeared more homogeneous but contained three smaller, previously unknown subpopulations.Conclusions Our study comprehensively characterized gene expression in individual glomerular cells and sets the stage for the dissection of glomerular function at the single-cell level in health and disease.


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.


2019 ◽  
Author(s):  
Chenling Xu ◽  
Romain Lopez ◽  
Edouard Mehlman ◽  
Jeffrey Regier ◽  
Michael I. Jordan ◽  
...  

AbstractAs single-cell transcriptomics becomes a mainstream technology, the natural next step is to integrate the accumulating data in order to achieve a common ontology of cell types and states. However, owing to various nuisance factors of variation, it is not straightforward how to compare gene expression levels across data sets and how to automatically assign cell type labels in a new data set based on existing annotations. In this manuscript, we demonstrate that our previously developed method, scVI, provides an effective and fully probabilistic approach for joint representation and analysis of cohorts of single-cell RNA-seq data sets, while accounting for uncertainty caused by biological and measurement noise. We also introduce single-cell ANnotation using Variational Inference (scANVI), a semi-supervised variant of scVI designed to leverage any available cell state annotations — for instance when only one data set in a cohort is annotated, or when only a few cells in a single data set can be labeled using marker genes. We demonstrate that scVI and scANVI compare favorably to the existing methods for data integration and cell state annotation in terms of accuracy, scalability, and adaptability to challenging settings such as a hierarchical structure of cell state labels. We further show that different from existing methods, scVI and scANVI represent the integrated datasets with a single generative model that can be directly used for any probabilistic decision making task, using differential expression as our case study. scVI and scANVI are available as open source software and can be readily used to facilitate cell state annotation and help ensure consistency and reproducibility across studies.


Sign in / Sign up

Export Citation Format

Share Document