scholarly journals EpiScanpy: integrated single-cell epigenomic analysis

2021 ◽  
Vol 12 (1) ◽  
Author(s):  
Anna Danese ◽  
Maria L. Richter ◽  
Kridsadakorn Chaichoompu ◽  
David S. Fischer ◽  
Fabian J. Theis ◽  
...  

AbstractEpiScanpy is a toolkit for the analysis of single-cell epigenomic data, namely single-cell DNA methylation and single-cell ATAC-seq data. To address the modality specific challenges from epigenomics data, epiScanpy quantifies the epigenome using multiple feature space constructions and builds a nearest neighbour graph using epigenomic distance between cells. EpiScanpy makes the many existing scRNA-seq workflows from scanpy available to large-scale single-cell data from other -omics modalities, including methods for common clustering, dimension reduction, cell type identification and trajectory learning techniques, as well as an atlas integration tool for scATAC-seq datasets. The toolkit also features numerous useful downstream functions, such as differential methylation and differential openness calling, mapping epigenomic features of interest to their nearest gene, or constructing gene activity matrices using chromatin openness. We successfully benchmark epiScanpy against other scATAC-seq analysis tools and show its outperformance at discriminating cell types.

2019 ◽  
Author(s):  
Anna Danese ◽  
Maria L. Richter ◽  
David S. Fischer ◽  
Fabian J. Theis ◽  
Maria Colomé-Tatché

ABSTRACTEpigenetic single-cell measurements reveal a layer of regulatory information not accessible to single-cell transcriptomics, however single-cell-omics analysis tools mainly focus on gene expression data. To address this issue, we present epiScanpy, a computational framework for the analysis of single-cell DNA methylation and single-cell ATAC-seq data. EpiScanpy makes the many existing RNA-seq workflows from scanpy available to large-scale single-cell data from other -omics modalities. We introduce and compare multiple feature space constructions for epigenetic data and show the feasibility of common clustering, dimension reduction and trajectory learning techniques. We benchmark epiScanpy by interrogating different single-cell brain mouse atlases of DNA methylation, ATAC-seq and transcriptomics. We find that differentially methylated and differentially open markers between cell clusters enrich transcriptome-based cell type labels by orthogonal epigenetic information.


2021 ◽  
Author(s):  
Jordan W. Squair ◽  
Michael A. Skinnider ◽  
Matthieu Gautier ◽  
Leonard J. Foster ◽  
Grégoire Courtine
Keyword(s):  

2021 ◽  
Vol 22 (S3) ◽  
Author(s):  
Yuanyuan Li ◽  
Ping Luo ◽  
Yi Lu ◽  
Fang-Xiang Wu

Abstract Background With the development of the technology of single-cell sequence, revealing homogeneity and heterogeneity between cells has become a new area of computational systems biology research. However, the clustering of cell types becomes more complex with the mutual penetration between different types of cells and the instability of gene expression. One way of overcoming this problem is to group similar, related single cells together by the means of various clustering analysis methods. Although some methods such as spectral clustering can do well in the identification of cell types, they only consider the similarities between cells and ignore the influence of dissimilarities on clustering results. This methodology may limit the performance of most of the conventional clustering algorithms for the identification of clusters, it needs to develop special methods for high-dimensional sparse categorical data. Results Inspired by the phenomenon that same type cells have similar gene expression patterns, but different types of cells evoke dissimilar gene expression patterns, we improve the existing spectral clustering method for clustering single-cell data that is based on both similarities and dissimilarities between cells. The method first measures the similarity/dissimilarity among cells, then constructs the incidence matrix by fusing similarity matrix with dissimilarity matrix, and, finally, uses the eigenvalues of the incidence matrix to perform dimensionality reduction and employs the K-means algorithm in the low dimensional space to achieve clustering. The proposed improved spectral clustering method is compared with the conventional spectral clustering method in recognizing cell types on several real single-cell RNA-seq datasets. Conclusions In summary, we show that adding intercellular dissimilarity can effectively improve accuracy and achieve robustness and that improved spectral clustering method outperforms the traditional spectral clustering method in grouping cells.


2020 ◽  
Vol 8 (Suppl 3) ◽  
pp. A520-A520
Author(s):  
Son Pham ◽  
Tri Le ◽  
Tan Phan ◽  
Minh Pham ◽  
Huy Nguyen ◽  
...  

BackgroundSingle-cell sequencing technology has opened an unprecedented ability to interrogate cancer. It reveals significant insights into the intratumoral heterogeneity, metastasis, therapeutic resistance, which facilitates target discovery and validation in cancer treatment. With rapid advancements in throughput and strategies, a particular immuno-oncology study can produce multi-omics profiles for several thousands of individual cells. This overflow of single-cell data poses formidable challenges, including standardizing data formats across studies, performing reanalysis for individual datasets and meta-analysis.MethodsN/AResultsWe present BioTuring Browser, an interactive platform for accessing and reanalyzing published single-cell omics data. The platform is currently hosting a curated database of more than 10 million cells from 247 projects, covering more than 120 immune cell types and subtypes, and 15 different cancer types. All data are processed and annotated with standardized labels of cell types, diseases, therapeutic responses, etc. to be instantly accessed and explored in a uniform visualization and analytics interface. Based on this massive curated database, BioTuring Browser supports searching similar expression profiles, querying a target across datasets and automatic cell type annotation. The platform supports single-cell RNA-seq, CITE-seq and TCR-seq data. BioTuring Browser is now available for download at www.bioturing.com.ConclusionsN/A


eLife ◽  
2021 ◽  
Vol 10 ◽  
Author(s):  
Alexander J Tarashansky ◽  
Jacob M Musser ◽  
Margarita Khariton ◽  
Pengyang Li ◽  
Detlev Arendt ◽  
...  

Comparing single-cell transcriptomic atlases from diverse organisms can elucidate the origins of cellular diversity and assist the annotation of new cell atlases. Yet, comparison between distant relatives is hindered by complex gene histories and diversifications in expression programs. Previously, we introduced the self-assembling manifold (SAM) algorithm to robustly reconstruct manifolds from single-cell data (Tarashansky et al., 2019). Here, we build on SAM to map cell atlas manifolds across species. This new method, SAMap, identifies homologous cell types with shared expression programs across distant species within phyla, even in complex examples where homologous tissues emerge from distinct germ layers. SAMap also finds many genes with more similar expression to their paralogs than their orthologs, suggesting paralog substitution may be more common in evolution than previously appreciated. Lastly, comparing species across animal phyla, spanning mouse to sponge, reveals ancient contractile and stem cell families, which may have arisen early in animal evolution.


2019 ◽  
Author(s):  
Michael Hagemann-Jensen ◽  
Christoph Ziegenhain ◽  
Ping Chen ◽  
Daniel Ramsköld ◽  
Gert-Jan Hendriks ◽  
...  

AbstractLarge-scale sequencing of RNAs from individual cells can reveal patterns of gene, isoform and allelic expression across cell types and states1. However, current single-cell RNA-sequencing (scRNA-seq) methods have limited ability to count RNAs at allele- and isoform resolution, and long-read sequencing techniques lack the depth required for large-scale applications across cells2,3. Here, we introduce Smart-seq3 that combines full-length transcriptome coverage with a 5’ unique molecular identifier (UMI) RNA counting strategy that enabled in silico reconstruction of thousands of RNA molecules per cell. Importantly, a large portion of counted and reconstructed RNA molecules could be directly assigned to specific isoforms and allelic origin, and we identified significant transcript isoform regulation in mouse strains and human cell types. Moreover, Smart-seq3 showed a dramatic increase in sensitivity and typically detected thousands more genes per cell than Smart-seq2. Altogether, we developed a short-read sequencing strategy for single-cell RNA counting at isoform and allele-resolution applicable to large-scale characterization of cell types and states across tissues and organisms.


Author(s):  
A. Nivaggioli ◽  
J. F. Hullo ◽  
G. Thibault

<p><strong>Abstract.</strong> Industrial companies often require complete inventories of their infrastructure. In many cases, a better inventory leads to a direct reduction of cost and uncertainty of engineering. While large scale panoramic surveys now allow these inventories to be performed remotely and reduce time on-site, the time and money required to visually segment the many types of components on thousands of high resolution panoramas can make the process infeasible. Recent studies have shown that deep learning techniques, namely deep neural networks, can accurately perform panoptic segmentation of <i>things</i> and <i>stuff</i> and hence be used to inventory the components of a picture. In order to train those deep architectures with specific industrial equipment, not available in public datasets, our approach uses an as-built 3D model of an industrial building to procedurally generate labels. Our results show that, despite the presence of errors during the generation of the dataset, our method is able to accurately perform panoptic segmentation on images of industrial scenes. In our testing, 80% of generated labels were correctly identified (non null intersection over union, i.e. true positive) by the panoptic segmentation, with great performance levels even for difficult classes, such as reflective heat insulators. We then visually investigated the 20% of true negative, and discovered that 80% were correctly segmented, but were counted as true negative because of errors in the dataset generation. Demonstrating this level of accuracy for panoptic segmentation on industrial panoramas for inventories also offers novel perspectives for 3D laser scan processing.</p>


2020 ◽  
Vol 2 (4) ◽  
Author(s):  
Kaikun Xie ◽  
Yu Huang ◽  
Feng Zeng ◽  
Zehua Liu ◽  
Ting Chen

Abstract Recent advancements in both single-cell RNA-sequencing technology and computational resources facilitate the study of cell types on global populations. Up to millions of cells can now be sequenced in one experiment; thus, accurate and efficient computational methods are needed to provide clustering and post-analysis of assigning putative and rare cell types. Here, we present a novel unsupervised deep learning clustering framework that is robust and highly scalable. To overcome the high level of noise, scAIDE first incorporates an autoencoder-imputation network with a distance-preserved embedding network (AIDE) to learn a good representation of data, and then applies a random projection hashing based k-means algorithm to accommodate the detection of rare cell types. We analyzed a 1.3 million neural cell dataset within 30 min, obtaining 64 clusters which were mapped to 19 putative cell types. In particular, we further identified three different neural stem cell developmental trajectories in these clusters. We also classified two subpopulations of malignant cells in a small glioblastoma dataset using scAIDE. We anticipate that scAIDE would provide a more in-depth understanding of cell development and diseases.


2020 ◽  
Vol 10 (1) ◽  
Author(s):  
Azam Peyvandipour ◽  
Adib Shafi ◽  
Nafiseh Saberian ◽  
Sorin Draghici
Keyword(s):  

2020 ◽  
Vol 36 (11) ◽  
pp. 3585-3587
Author(s):  
Lin Wang ◽  
Francisca Catalan ◽  
Karin Shamardani ◽  
Husam Babikir ◽  
Aaron Diaz

Abstract Summary Single-cell data are being generated at an accelerating pace. How best to project data across single-cell atlases is an open problem. We developed a boosted learner that overcomes the greatest challenge with status quo classifiers: low sensitivity, especially when dealing with rare cell types. By comparing novel and published data from distinct scRNA-seq modalities that were acquired from the same tissues, we show that this approach preserves cell-type labels when mapping across diverse platforms. Availability and implementation https://github.com/diazlab/ELSA Contact [email protected] Supplementary information Supplementary data are available at Bioinformatics online.


Sign in / Sign up

Export Citation Format

Share Document