scholarly journals scAIDE: clustering of large-scale single-cell RNA-seq data reveals putative and rare cell types

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.

2018 ◽  
Author(s):  
Xianwen Ren ◽  
Liangtao Zheng ◽  
Zemin Zhang

ABSTRACTClustering is a prevalent analytical means to analyze single cell RNA sequencing data but the rapidly expanding data volume can make this process computational challenging. New methods for both accurate and efficient clustering are of pressing needs. Here we proposed a new clustering framework based on random projection and feature construction for large scale single-cell RNA sequencing data, which greatly improves clustering accuracy, robustness and computational efficacy for various state-of-the-art algorithms benchmarked on multiple real datasets. On a dataset with 68,578 human blood cells, our method reached 20% improvements for clustering accuracy and 50-fold acceleration but only consumed 66% memory usage compared to the widely-used software package SC3. Compared to k-means, the accuracy improvement can reach 3-fold depending on the concrete dataset. An R implementation of the framework is available from https://github.com/Japrin/sscClust.


2020 ◽  
Author(s):  
Van Hoan Do ◽  
Francisca Rojas Ringeling ◽  
Stefan Canzar

AbstractA fundamental task in single-cell RNA-seq (scRNA-seq) analysis is the identification of transcriptionally distinct groups of cells. Numerous methods have been proposed for this problem, with a recent focus on methods for the cluster analysis of ultra-large scRNA-seq data sets produced by droplet-based sequencing technologies. Most existing methods rely on a sampling step to bridge the gap between algorithm scalability and volume of the data. Ignoring large parts of the data, however, often yields inaccurate groupings of cells and risks overlooking rare cell types. We propose method Specter that adopts and extends recent algorithmic advances in (fast) spectral clustering. In contrast to methods that cluster a (random) subsample of the data, we adopt the idea of landmarks that are used to create a sparse representation of the full data from which a spectral embedding can then be computed in linear time. We exploit Specter’s speed in a cluster ensemble scheme that achieves a substantial improvement in accuracy over existing methods and that is sensitive to rare cell types. Its linear time complexity allows Specter to scale to millions of cells and leads to fast computation times in practice. Furthermore, on CITE-seq data that simultaneously measures gene and protein marker expression we demonstrate that Specter is able to utilize multimodal omics measurements to resolve subtle transcriptomic differences between subpopulations of cells. Specter is open source and available at https://github.com/canzarlab/Specter.


2022 ◽  
Vol 23 (1) ◽  
Author(s):  
Huijian Feng ◽  
Lihui Lin ◽  
Jiekai Chen

Abstract Background Single-cell RNA sequencing is becoming a powerful tool to identify cell states, reconstruct developmental trajectories, and deconvolute spatial expression. The rapid development of computational methods promotes the insight of heterogeneous single-cell data. An increasing number of tools have been provided for biological analysts, of which two programming languages- R and Python are widely used among researchers. R and Python are complementary, as many methods are implemented specifically in R or Python. However, the different platforms immediately caused the data sharing and transformation problem, especially for Scanpy, Seurat, and SingleCellExperiemnt. Currently, there is no efficient and user-friendly software to perform data transformation of single-cell omics between platforms, which makes users spend unbearable time on data Input and Output (IO), significantly reducing the efficiency of data analysis. Results We developed scDIOR for single-cell data transformation between platforms of R and Python based on Hierarchical Data Format Version 5 (HDF5). We have created a data IO ecosystem between three R packages (Seurat, SingleCellExperiment, Monocle) and a Python package (Scanpy). Importantly, scDIOR accommodates a variety of data types across programming languages and platforms in an ultrafast way, including single-cell RNA-seq and spatial resolved transcriptomics data, using only a few codes in IDE or command line interface. For large scale datasets, users can partially load the needed information, e.g., cell annotation without the gene expression matrices. scDIOR connects the analytical tasks of different platforms, which makes it easy to compare the performance of algorithms between them. Conclusions scDIOR contains two modules, dior in R and diopy in Python. scDIOR is a versatile and user-friendly tool that implements single-cell data transformation between R and Python rapidly and stably. The software is freely accessible at https://github.com/JiekaiLab/scDIOR.


2018 ◽  
Author(s):  
Chieh Lin ◽  
Ziv Bar-Joseph

AbstractMotivationMethods for reconstructing developmental trajectories from time series single cell RNA-Seq (scRNA-Seq) data can be largely divided into two categories. The first, often referred to as pseudotime ordering methods, are deterministic and rely on dimensionality reduction followed by an ordering step. The second learns a probabilistic branching model to represent the developmental process. While both types have been successful, each suffers from shortcomings that can impact their accuracy.ResultsWe developed a new method based on continuous state HMMs (CSHMMs) for representing and modeling time series scRNA-Seq data. We define the CSHMM model and provide efficient learning and inference algorithms which allow the method to determine both the structure of the branching process and the assignment of cells to these branches. Analyzing several developmental single cell datasets we show that the CSHMM method accurately infers branching topology and correctly and continuously assign cells to paths, improving upon prior methods proposed for this task. Analysis of genes based on the continuous cell assignment identifies known and novel markers for different cell types.AvailabilitySoftware and Supporting website: www.andrew.cmu.edu/user/chiehll/CSHMM/[email protected] informationSupplementary data are available at Bioinformatics online.


2017 ◽  
Author(s):  
Megan Crow ◽  
Anirban Paul ◽  
Sara Ballouz ◽  
Z. Josh Huang ◽  
Jesse Gillis

AbstractSingle cell RNA-sequencing technology (scRNA-seq) provides a new avenue to discover and characterize cell types, but the experiment-specific technical biases and analytic variability inherent to current pipelines may undermine the replicability of these studies. Meta-analysis of rapidly accumulating data is further hampered by the use of ad hoc naming conventions. Here we demonstrate our replication framework, MetaNeighbor, that allows researchers to quantify the degree to which cell types replicate across datasets, and to rapidly identify clusters with high similarity for further testing. We first measure the replicability of neuronal identity by comparing more than 13 thousand individual scRNA-seq transcriptomes, sampling with high specificity from within the data to define a range of robust practices. We then assess cross-dataset evidence for novel cortical interneuron subtypes identified by scRNA-seq and find that 24/45 cortical interneuron subtypes have evidence of replication in at least one other study. Identifying these putative replicates allows us to re-analyze the data for differential expression and provide lists of robust candidate marker genes. Across tasks we find that large sets of variably expressed genes can identify replicable cell types and subtypes with high accuracy, suggesting a general route forward for large-scale evaluation of scRNA-seq data.


2020 ◽  
Author(s):  
Bo Liu ◽  
Fang-Xiang Wu ◽  
Xiufen Zou

ABSTRACTThe Human Cell Atlas (HCA) is a large project that aims to identify all cell types in the human body. The dimension reduction and clustering for identification of cell types from single-cell RNA-sequencing (scRNA-seq) data have become foundational approaches to HCA. The major challenges of current computational analyses are of poor performance on large scale data and sensitive to initial data. We present a new ensemble framework called Adaptive Slice KNNs (scASK) to address the challenges for analysing scRNA-seq data with high dimensionality. scASK consists of three innovational modules, called DAS (Data Adaptive Slicing), MCS (Meta Classifiers Selecting) and EMS (Ensemble Mode Switching), respectively, which facilitate scASK to approximate a bias-variance tradeoff beyond classification. Thirteen real scRNA-seq datasets are used to evaluate the performance of scASK. Compared with five popular classification algorithms, our experimental results indicate that scASK achieves the best accuracy and robustness among all competing methods. In conclusion, adaptive slicing is an effective structural reduction procedure, and meanwhile scASK provides novel and robust ensemble framework especially for classifying cell types based on scRNA-seq data. scASK is publically available at https://github.com/liubo2358/scASKcmd.


2022 ◽  
Author(s):  
Chenfei Wang ◽  
Pengfei Ren ◽  
Xiaoying Shi ◽  
Xin Dong ◽  
Zhiguang Yu ◽  
...  

Abstract The rapid accumulation of single-cell RNA-seq data has provided rich resources to characterize various human cell types. Cell type annotation is the critical step in analyzing single-cell RNA-seq data. However, accurate cell type annotation based on public references is challenging due to the inconsistent annotations, batch effects, and poor characterization of rare cell types. Here, we introduce SELINA (single cELl identity NAvigator), an integrative annotation transferring framework for automatic cell type annotation. SELINA optimizes the annotation for minority cell types by synthetic minority over-sampling, removes batch effects among reference datasets using a multiple-adversarial domain adaptation network (MADA), and fits the query data with reference data using an autoencoder. Finally, SELINA affords a comprehensive and uniform reference atlas with 1.7 million cells covering 230 major human cell types. We demonstrated the robustness and superiority of SELINA in most human tissues compared to existing methods. SELINA provided a one-stop solution for human single- cell RNA-seq data annotation with the potential to extend for other species.


2019 ◽  
Vol 36 (8) ◽  
pp. 2474-2485 ◽  
Author(s):  
Zhanying Feng ◽  
Xianwen Ren ◽  
Yuan Fang ◽  
Yining Yin ◽  
Chutian Huang ◽  
...  

Abstract Motivation Single cell RNA-seq data offers us new resource and resolution to study cell type identity and its conversion. However, data analyses are challenging in dealing with noise, sparsity and poor annotation at single cell resolution. Detecting cell-type-indicative markers is promising to help denoising, clustering and cell type annotation. Results We developed a new method, scTIM, to reveal cell-type-indicative markers. scTIM is based on a multi-objective optimization framework to simultaneously maximize gene specificity by considering gene-cell relationship, maximize gene’s ability to reconstruct cell–cell relationship and minimize gene redundancy by considering gene–gene relationship. Furthermore, consensus optimization is introduced for robust solution. Experimental results on three diverse single cell RNA-seq datasets show scTIM’s advantages in identifying cell types (clustering), annotating cell types and reconstructing cell development trajectory. Applying scTIM to the large-scale mouse cell atlas data identifies critical markers for 15 tissues as ‘mouse cell marker atlas’, which allows us to investigate identities of different tissues and subtle cell types within a tissue. scTIM will serve as a useful method for single cell RNA-seq data mining. Availability and implementation scTIM is freely available at https://github.com/Frank-Orwell/scTIM. Supplementary information Supplementary data are available at Bioinformatics online.


2018 ◽  
Author(s):  
Amir Alavi ◽  
Matthew Ruffalo ◽  
Aiyappa Parvangada ◽  
Zhilin Huang ◽  
Ziv Bar-Joseph

SummarySingle cell RNA-Seq (scRNA-seq) studies often profile upward of thousands of cells in heterogeneous environments. Current methods for characterizing cells perform unsupervised analysis followed by assignment using a small set of known marker genes. Such approaches are limited to a few, well characterized cell types. To enable large scale supervised characterization we developed an automated pipeline to download, process, and annotate publicly available scRNA-seq datasets. We extended supervised neural networks to obtain efficient and accurate representations for scRNA-seq data. We applied our pipeline to analyze data from over 500 different studies with over 300 unique cell types and show that supervised methods greatly outperform unsupervised methods for cell type identification. A case study of neural degeneration data highlights the ability of these methods to identify differences between cell type distributions in healthy and diseased mice. We implemented a web server that compares new datasets to collected data employing fast matching methods in order to determine cell types, key genes, similar prior studies, and more.


2018 ◽  
Author(s):  
Shibiao Wan ◽  
Junil Kim ◽  
Kyoung Jae Won

ABSTRACTTo process large-scale single-cell RNA-sequencing (scRNA-seq) data effectively without excessive distortion during dimension reduction, we present SHARP, an ensemble random projection-based algorithm which is scalable to clustering 10 million cells. Comprehensive benchmarking tests on 17 public scRNA-seq datasets demonstrate that SHARP outperforms existing methods in terms of speed and accuracy. Particularly, for large-size datasets (>40,000 cells), SHARP’s running speed far excels other competitors while maintaining high clustering accuracy and robustness. To the best of our knowledge, SHARP is the only R-based tool that is scalable to clustering scRNA-seq data with 10 million cells.


Sign in / Sign up

Export Citation Format

Share Document