首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 0 毫秒
1.
2.
3.

Background

Human cancers are complex ecosystems composed of cells with distinct molecular signatures. Such intratumoral heterogeneity poses a major challenge to cancer diagnosis and treatment. Recent advancements of single-cell techniques such as scRNA-seq have brought unprecedented insights into cellular heterogeneity. Subsequently, a challenging computational problem is to cluster high dimensional noisy datasets with substantially fewer cells than the number of genes.

Methods

In this paper, we introduced a consensus clustering framework conCluster, for cancer subtype identification from single-cell RNA-seq data. Using an ensemble strategy, conCluster fuses multiple basic partitions to consensus clusters.

Results

Applied to real cancer scRNA-seq datasets, conCluster can more accurately detect cancer subtypes than the widely used scRNA-seq clustering methods. Further, we conducted co-expression network analysis for the identified melanoma subtypes.

Conclusions

Our analysis demonstrates that these subtypes exhibit distinct gene co-expression networks and significant gene sets with different functional enrichment.
  相似文献   

4.
Annotation of cells in single-cell clustering requires a homogeneous grouping of cell populations. There are various issues in single cell sequencing that effect homogeneous grouping (clustering) of cells, such as small amount of starting RNA, limited per-cell sequenced reads, cell-to-cell variability due to cell-cycle, cellular morphology, and variable reagent concentrations. Moreover, single cell data is susceptible to technical noise, which affects the quality of genes (or features) selected/extracted prior to clustering.Here we introduce sc-CGconv (copula based graph convolution network for single clustering), a stepwise robust unsupervised feature extraction and clustering approach that formulates and aggregates cell–cell relationships using copula correlation (Ccor), followed by a graph convolution network based clustering approach. sc-CGconv formulates a cell-cell graph using Ccor that is learned by a graph-based artificial intelligence model, graph convolution network. The learned representation (low dimensional embedding) is utilized for cell clustering. sc-CGconv features the following advantages. a. sc-CGconv works with substantially smaller sample sizes to identify homogeneous clusters. b. sc-CGconv can model the expression co-variability of a large number of genes, thereby outperforming state-of-the-art gene selection/extraction methods for clustering. c. sc-CGconv preserves the cell-to-cell variability within the selected gene set by constructing a cell-cell graph through copula correlation measure. d. sc-CGconv provides a topology-preserving embedding of cells in low dimensional space.  相似文献   

5.
《Genomics》2021,113(3):1308-1324
Single-cell RNA sequencing (scRNA-seq) is a powerful technology that is capable of generating gene expression data at the resolution of individual cell. The scRNA-seq data is characterized by the presence of dropout events, which severely bias the results if they remain unaddressed. There are limited Differential Expression (DE) approaches which consider the biological processes, which lead to dropout events, in the modeling process. So, we develop, SwarnSeq, an improved method for DE, and other downstream analysis that considers the molecular capture process in scRNA-seq data modeling. The performance of the proposed method is benchmarked with 11 existing methods on 10 different real scRNA-seq datasets under three comparison settings. We demonstrate that SwarnSeq method has improved performance over the 11 existing methods. This improvement is consistently observed across several public scRNA-seq datasets generated using different scRNA-seq protocols. The external spike-ins data can be used in the SwarnSeq method to enhance its performance.Availability and implementationThe method is implemented as a publicly available R package available at https://github.com/sam-uofl/SwarnSeq.  相似文献   

6.
As the cost of single-cell RNA-seq experiments has decreased, an increasing number of datasets are now available. Combining newly generated and publicly accessible datasets is challenging due to non-biological signals, commonly known as batch effects. Although there are several computational methods available that can remove batch effects, evaluating which method performs best is not straightforward. Here, we present BatchBench (https://github.com/cellgeni/batchbench), a modular and flexible pipeline for comparing batch correction methods for single-cell RNA-seq data. We apply BatchBench to eight methods, highlighting their methodological differences and assess their performance and computational requirements through a compendium of well-studied datasets. This systematic comparison guides users in the choice of batch correction tool, and the pipeline makes it easy to evaluate other datasets.  相似文献   

7.
The single-cell RNA sequencing (scRNA-seq) technologies obtain gene expression at single-cell resolution and provide a tool for exploring cell heterogeneity and cell types. As the low amount of extracted mRNA copies per cell, scRNA-seq data exhibit a large number of dropouts, which hinders the downstream analysis of the scRNA-seq data. We propose a statistical method, SDImpute (Single-cell RNA-seq Dropout Imputation), to implement block imputation for dropout events in scRNA-seq data. SDImpute automatically identifies the dropout events based on the gene expression levels and the variations of gene expression across similar cells and similar genes, and it implements block imputation for dropouts by utilizing gene expression unaffected by dropouts from similar cells. In the experiments, the results of the simulated datasets and real datasets suggest that SDImpute is an effective tool to recover the data and preserve the heterogeneity of gene expression across cells. Compared with the state-of-the-art imputation methods, SDImpute improves the accuracy of the downstream analysis including clustering, visualization, and differential expression analysis.  相似文献   

8.
Sun W 《Biometrics》2012,68(1):1-11
RNA-seq may replace gene expression microarrays in the near future. Using RNA-seq, the expression of a gene can be estimated using the total number of sequence reads mapped to that gene, known as the total read count (TReC). Traditional expression quantitative trait locus (eQTL) mapping methods, such as linear regression, can be applied to TReC measurements after they are properly normalized. In this article, we show that eQTL mapping, by directly modeling TReC using discrete distributions, has higher statistical power than the two-step approach: data normalization followed by linear regression. In addition, RNA-seq provides information on allele-specific expression (ASE) that is not available from microarrays. By combining the information from TReC and ASE, we can computationally distinguish cis- and trans-eQTL and further improve the power of cis-eQTL mapping. Both simulation and real data studies confirm the improved power of our new methods. We also discuss the design issues of RNA-seq experiments. Specifically, we show that by combining TReC and ASE measurements, it is possible to minimize cost and retain the statistical power of cis-eQTL mapping by reducing sample size while increasing the number of sequence reads per sample. In addition to RNA-seq data, our method can also be employed to study the genetic basis of other types of sequencing data, such as chromatin immunoprecipitation followed by DNA sequencing data. In this article, we focus on eQTL mapping of a single gene using the association-based method. However, our method establishes a statistical framework for future developments of eQTL mapping methods using RNA-seq data (e.g., linkage-based eQTL mapping), and the joint study of multiple genetic markers and/or multiple genes.  相似文献   

9.
10.
11.

Background

Gene signatures are important to represent the molecular changes in the disease genomes or the cells in specific conditions, and have been often used to separate samples into different groups for better research or clinical treatment. While many methods and applications have been available in literature, there still lack powerful ones that can take account of the complex data and detect the most informative signatures.

Methods

In this article, we present a new framework for identifying gene signatures using Pareto-optimal cluster size identification for RNA-seq data. We first performed pre-filtering steps and normalization, then utilized the empirical Bayes test in Limma package to identify the differentially expressed genes (DEGs). Next, we used a multi-objective optimization technique, “Multi-objective optimization for collecting cluster alternatives” (MOCCA in R package) on these DEGs to find Pareto-optimal cluster size, and then applied k-means clustering to the RNA-seq data based on the optimal cluster size. The best cluster was obtained through computing the average Spearman’s Correlation Score among all the genes in pair-wise manner belonging to the module. The best cluster is treated as the signature for the respective disease or cellular condition.

Results

We applied our framework to a cervical cancer RNA-seq dataset, which included 253 squamous cell carcinoma (SCC) samples and 22 adenocarcinoma (ADENO) samples. We identified a total of 582 DEGs by Limma analysis of SCC versus ADENO samples. Among them, 260 are up-regulated genes and 322 are down-regulated genes. Using MOCCA, we obtained seven Pareto-optimal clusters. The best cluster has a total of 35 DEGs consisting of all-upregulated genes. For validation, we ran PAMR (prediction analysis for microarrays) classifier on the selected best cluster, and assessed the classification performance. Our evaluation, measured by sensitivity, specificity, precision, and accuracy, showed high confidence.

Conclusions

Our framework identified a multi-objective based cluster that is treated as a signature that can classify the disease and control group of samples with higher classification performance (accuracy 0.935) for the corresponding disease. Our method is useful to find signature for any RNA-seq or microarray data.
  相似文献   

12.
The standard analysis pipeline for single-cell RNA-seq data consists of sequential steps initiated by clustering the cells. An innate limitation of this pipeline is that an imperfect clustering result can irreversibly affect the succeeding steps. For example, there can be cell types not well distinguished by clustering because they largely share the global structure, such as the anterior primitive streak and mid primitive streak cells. If one searches differentially expressed genes (DEGs) solely based on clustering, marker genes for distinguishing these types will be missed. Moreover, clustering depends on many parameters and can often be subjective to manual decisions. To overcome these limitations, we propose MarcoPolo, a method that identifies informative DEGs independently of prior clustering. MarcoPolo sorts out genes by evaluating if the distributions are bimodal, if similar expression patterns are observed in other genes, and if the expressing cells are proximal in a low-dimensional space. Using real datasets with FACS-purified cell labels, we demonstrate that MarcoPolo recovers marker genes better than competing methods. Notably, MarcoPolo finds key genes that can distinguish cell types that are not distinguishable by the standard clustering. MarcoPolo is built in a convenient software package that provides analysis results in an HTML file.  相似文献   

13.
A large number of computational methods have been developed for analyzing differential gene expression in RNA-seq data. We describe a comprehensive evaluation of common methods using the SEQC benchmark dataset and ENCODE data. We consider a number of key features, including normalization, accuracy of differential expression detection and differential expression analysis when one condition has no detectable expression. We find significant differences among the methods, but note that array-based methods adapted to RNA-seq data perform comparably to methods designed for RNA-seq. Our results demonstrate that increasing the number of replicate samples significantly improves detection power over increased sequencing depth.  相似文献   

14.
15.
《Genomics》2022,114(2):110264
Cancer is one of the major causes of human death per year. In recent years, cancer identification and classification using machine learning have gained momentum due to the availability of high throughput sequencing data. Using RNA-seq, cancer research is blooming day by day and new insights of cancer and related treatments are coming into light. In this paper, we propose PanClassif, a method that requires a very few and effective genes to detect cancer from RNA-seq data and is able to provide performance gain in several wide range machine learning classifiers. We have taken 22 types of cancer samples from The Cancer Genome Atlas (TCGA) having 8287 cancer samples and 680 normal samples. Firstly, PanClassif uses k-Nearest Neighbour (k-NN) smoothing to smooth the samples to handle noise in the data. Then effective genes are selected by Anova based test. For balancing the train data, PanClassif applies an oversampling method, SMOTE. We have performed comprehensive experiments on the datasets using several classification algorithms. Experimental results shows that PanClassif outperform existing state-of-the-art methods available and shows consistent performance for two single cell RNA-seq datasets taken from Gene Expression Omnibus (GEO). PanClassif improves performances of a wide variety of classifiers for both binary cancer prediction and multi-class cancer classification. PanClassif is available as a python package (https://pypi.org/project/panclassif/). All the source code and materials of PanClassif are available at https://github.com/Zwei-inc/panclassif.  相似文献   

16.
17.
18.
19.
Single-cell Hi-C (scHi-C) sequencing technologies allow us to investigate three-dimensional chromatin organization at the single-cell level. However, we still need computational tools to deal with the sparsity of the contact maps from single cells and embed single cells in a lower-dimensional Euclidean space. This embedding helps us understand relationships between the cells in different dimensions, such as cell-cycle dynamics and cell differentiation. We present an open-source computational toolbox, scHiCTools, for analyzing single-cell Hi-C data comprehensively and efficiently. The toolbox provides two methods for screening single cells, three common methods for smoothing scHi-C data, three efficient methods for calculating the pairwise similarity of cells, three methods for embedding single cells, three methods for clustering cells, and a build-in function to visualize the cells embedding in a two-dimensional or three-dimensional plot. scHiCTools, written in Python3, is compatible with different platforms, including Linux, macOS, and Windows.  相似文献   

20.
设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号