Single-Cell Rna-Seq Reveals Cellular Heterogeneity of Pluripotency Transition and X-Chromosome Dynamics During Early Postimplantation Mouse Development

2018 ◽  
Author(s):  
Shangli Cheng ◽  
Yu Pei ◽  
Liqun He ◽  
Guangdun Peng ◽  
Björn Reinius ◽  
...  
Cell Reports ◽  
2019 ◽  
Vol 26 (10) ◽  
pp. 2593-2607.e3 ◽  
Author(s):  
Shangli Cheng ◽  
Yu Pei ◽  
Liqun He ◽  
Guangdun Peng ◽  
Björn Reinius ◽  
...  

Cell ◽  
2016 ◽  
Vol 165 (4) ◽  
pp. 1012-1026 ◽  
Author(s):  
Sophie Petropoulos ◽  
Daniel Edsgärd ◽  
Björn Reinius ◽  
Qiaolin Deng ◽  
Sarita Pauliina Panula ◽  
...  

Cell ◽  
2016 ◽  
Vol 167 (1) ◽  
pp. 285 ◽  
Author(s):  
Sophie Petropoulos ◽  
Daniel Edsgärd ◽  
Björn Reinius ◽  
Qiaolin Deng ◽  
Sarita Pauliina Panula ◽  
...  

Author(s):  
Congting Ye ◽  
Qian Zhou ◽  
Xiaohui Wu ◽  
Chen Yu ◽  
Guoli Ji ◽  
...  

Abstract Motivation Alternative polyadenylation (APA) plays a key post-transcriptional regulatory role in mRNA stability and functions in eukaryotes. Single cell RNA-seq (scRNA-seq) is a powerful tool to discover cellular heterogeneity at gene expression level. Given 3′ enriched strategy in library construction, the most commonly used scRNA-seq protocol—10× Genomics enables us to improve the study resolution of APA to the single cell level. However, currently there is no computational tool available for investigating APA profiles from scRNA-seq data. Results Here, we present a package scDAPA for detecting and visualizing dynamic APA from scRNA-seq data. Taking bam/sam files and cell cluster labels as inputs, scDAPA detects APA dynamics using a histogram-based method and the Wilcoxon rank-sum test, and visualizes candidate genes with dynamic APA. Benchmarking results demonstrated that scDAPA can effectively identify genes with dynamic APA among different cell groups from scRNA-seq data. Availability and implementation The scDAPA package is implemented in Shell and R, and is freely available at https://scdapa.sourceforge.io. Contact [email protected] Supplementary information Supplementary data are available at Bioinformatics online.


2020 ◽  
Vol 36 (15) ◽  
pp. 4233-4239
Author(s):  
Di Ran ◽  
Shanshan Zhang ◽  
Nicholas Lytal ◽  
Lingling An

Abstract Motivation Single-cell RNA-sequencing (scRNA-seq) has become an important tool to unravel cellular heterogeneity, discover new cell (sub)types, and understand cell development at single-cell resolution. However, one major challenge to scRNA-seq research is the presence of ‘drop-out’ events, which usually is due to extremely low mRNA input or the stochastic nature of gene expression. In this article, we present a novel single-cell RNA-seq drop-out correction (scDoc) method, imputing drop-out events by borrowing information for the same gene from highly similar cells. Results scDoc is the first method that directly involves drop-out information to accounting for cell-to-cell similarity estimation, which is crucial in scRNA-seq drop-out imputation but has not been appropriately examined. We evaluated the performance of scDoc using both simulated data and real scRNA-seq studies. Results show that scDoc outperforms the existing imputation methods in reference to data visualization, cell subpopulation identification and differential expression detection in scRNA-seq data. Availability and implementation R code is available at https://github.com/anlingUA/scDoc. Supplementary information Supplementary data are available at Bioinformatics online.


2020 ◽  
Author(s):  
Jixing Zhong ◽  
Gen Tang ◽  
Jiacheng Zhu ◽  
Xin Qiu ◽  
Weiying Wu ◽  
...  

AbstractParkinson’s disease (PD) is a neurodegenerative disease leading to the impairment of execution of movement. PD pathogenesis has been largely investigated, but either restricted in bulk level or at certain cell types, which failed to capture cellular heterogeneity and intrinsic interplays among distinct cell types. To overcome this, we applied single-nucleus RNA-seq and single cell ATAC-seq on cerebellum, midbrain and striatum of PD mouse and matched control. With 74,493 cells in total, we comprehensively depicted the dysfunctions under PD pathology covering proteostasis, neuroinflammation, calcium homeostasis and extracellular neurotransmitter homeostasis. Besides, by multi-omics approach, we identified putative biomarkers for early stage of PD, based on the relationships between transcriptomic and epigenetic profiles. We located certain cell types that primarily contribute to PD early pathology, narrowing the gap between genotypes and phenotypes. Taken together, our study provides a valuable resource to dissect the molecular mechanism of PD pathogenesis at single cell level, which could facilitate the development of novel methods regarding diagnosis, monitoring and practical therapies against PD at early stage.


2020 ◽  
Author(s):  
Matthew N. Bernstein ◽  
Zijian Ni ◽  
Michael Collins ◽  
Mark E. Burkard ◽  
Christina Kendziorski ◽  
...  

AbstractBackgroundSingle-cell RNA-seq (scRNA-seq) enables the profiling of genome-wide gene expression at the single-cell level and in so doing facilitates insight into and information about cellular heterogeneity within a tissue. Perhaps nowhere is this more important than in cancer, where tumor and tumor microenvironment heterogeneity directly impact development, maintenance, and progression of disease. While publicly available scRNA-seq cancer datasets offer unprecedented opportunity to better understand the mechanisms underlying tumor progression, metastasis, drug resistance, and immune evasion, much of the available information has been underutilized, in part, due to the lack of tools available for aggregating and analysing these data.ResultsWe present CHARacterizing Tumor Subpopulations (CHARTS), a computational pipeline and web application for analyzing, characterizing, and integrating publicly available scRNA-seq cancer datasets. CHARTS enables the exploration of individual gene expression, cell type, malignancy-status, differentially expressed genes, and gene set enrichment results in subpopulations of cells across multiple tumors and datasets.ConclusionCHARTS is an easy to use, comprehensive platform for exploring single-cell subpopulations within tumors across the ever-growing collection of public scRNA-seq cancer datasets. CHARTS is freely available at charts.morgridge.org.


2018 ◽  
Author(s):  
Xuran Wang ◽  
Jihwan Park ◽  
Katalin Susztak ◽  
Nancy R. Zhang ◽  
Mingyao Li

AbstractWe present MuSiC, a method that utilizes cell-type specific gene expression from single-cell RNA sequencing (RNA-seq) data to characterize cell type compositions from bulk RNA-seq data in complex tissues. When applied to pancreatic islet and whole kidney expression data in human, mouse, and rats, MuSiC outperformed existing methods, especially for tissues with closely related cell types. MuSiC enables characterization of cellular heterogeneity of complex tissues for identification of disease mechanisms.


Sign in / Sign up

Export Citation Format

Share Document