• Title/Summary/Keyword: single-cell RNA-seq

Search Result 23, Processing Time 0.022 seconds

Integration of Single-Cell RNA-Seq Datasets: A Review of Computational Methods

  • Yeonjae Ryu;Geun Hee Han;Eunsoo Jung;Daehee Hwang
    • Molecules and Cells
    • /
    • v.46 no.2
    • /
    • pp.106-119
    • /
    • 2023
  • With the increased number of single-cell RNA sequencing (scRNA-seq) datasets in public repositories, integrative analysis of multiple scRNA-seq datasets has become commonplace. Batch effects among different datasets are inevitable because of differences in cell isolation and handling protocols, library preparation technology, and sequencing platforms. To remove these batch effects for effective integration of multiple scRNA-seq datasets, a number of methodologies have been developed based on diverse concepts and approaches. These methods have proven useful for examining whether cellular features, such as cell subpopulations and marker genes, identified from a certain dataset, are consistently present, or whether their condition-dependent variations, such as increases in cell subpopulations in particular disease-related conditions, are consistently observed in different datasets generated under similar or distinct conditions. In this review, we summarize the concepts and approaches of the integration methods and their pros and cons as has been reported in previous literature.

Dissecting Cellular Heterogeneity Using Single-Cell RNA Sequencing

  • Choi, Yoon Ha;Kim, Jong Kyoung
    • Molecules and Cells
    • /
    • v.42 no.3
    • /
    • pp.189-199
    • /
    • 2019
  • Cell-to-cell variability in gene expression exists even in a homogeneous population of cells. Dissecting such cellular heterogeneity within a biological system is a prerequisite for understanding how a biological system is developed, homeostatically regulated, and responds to external perturbations. Single-cell RNA sequencing (scRNA-seq) allows the quantitative and unbiased characterization of cellular heterogeneity by providing genome-wide molecular profiles from tens of thousands of individual cells. A major question in analyzing scRNA-seq data is how to account for the observed cell-to-cell variability. In this review, we provide an overview of scRNA-seq protocols, computational approaches for dissecting cellular heterogeneity, and future directions of single-cell transcriptomic analysis.

Variational Autoencoder Based Dimension Reduction and Clustering for Single-Cell RNA-seq Gene Expression (단일세포 RNA-SEQ의 유전자 발현 군집화를 위한 변이 자동인코더 기반의 차원감소와 군집화)

  • Chi, Sang-Mun
    • Journal of the Korea Institute of Information and Communication Engineering
    • /
    • v.25 no.11
    • /
    • pp.1512-1518
    • /
    • 2021
  • Since single cell RNA sequencing provides the expression profiles of individual cells, it provides higher cellular differential resolution than traditional bulk RNA sequencing. Using these single cell RNA sequencing data, clustering analysis is generally conducted to find cell types and understand high level biological processes. In order to effectively process the high-dimensional single cell RNA sequencing data fir the clustering analysis, this paper uses a variational autoencoder to transform a high dimensional data space into a lower dimensional latent space, expecting to produce a latent space that can give more accurate clustering results. By clustering the features in the transformed latent space, we compare the performance of various classical clustering methods for single cell RNA sequencing data. Experimental results demonstrate that the proposed framework outperforms many state-of-the-art methods under various clustering performance metrics.

Single-cell and spatial transcriptomics approaches of cardiovascular development and disease

  • Roth, Robert;Kim, Soochi;Kim, Jeesu;Rhee, Siyeon
    • BMB Reports
    • /
    • v.53 no.8
    • /
    • pp.393-399
    • /
    • 2020
  • Recent advancements in the resolution and throughput of single-cell analyses, including single-cell RNA sequencing (scRNA-seq), have achieved significant progress in biomedical research in the last decade. These techniques have been used to understand cellular heterogeneity by identifying many rare and novel cell types and characterizing subpopulations of cells that make up organs and tissues. Analysis across various datasets can elucidate temporal patterning in gene expression and developmental cues and is also employed to examine the response of cells to acute injury, damage, or disruption. Specifically, scRNA-seq and spatially resolved transcriptomics have been used to describe the identity of novel or rare cell subpopulations and transcriptional variations that are related to normal and pathological conditions in mammalian models and human tissues. These applications have critically contributed to advance basic cardiovascular research in the past decade by identifying novel cell types implicated in development and disease. In this review, we describe current scRNA-seq technologies and how current scRNA-seq and spatial transcriptomic (ST) techniques have advanced our understanding of cardiovascular development and disease.

Identification of ERBB pathway-activated cells in triple-negative breast cancer

  • Cho, Soo Young
    • Genomics & Informatics
    • /
    • v.17 no.1
    • /
    • pp.3.1-3.4
    • /
    • 2019
  • Intratumor heterogeneity within a single tumor mass is one of the hallmarks of malignancy and has been reported in various tumor types. The molecular characterization of intratumor heterogeneity in breast cancer is a significant challenge for effective treatment. Using single-cell RNA sequencing (RNA-seq) data from a public resource, an ERBB pathway activated triple-negative cell population was identified. The differential expression of three subtyping marker genes (ERBB2, ESR1, and PGR) was not changed in the bulk RNA-seq data, but the single-cell transcriptomes showed intratumor heterogeneity. This result shows that ERBB signaling is activated using an indirect route and that the molecular subtype is changed on a single-cell level. Our data propose a different view on breast cancer subtypes, clarifying much confusion in this field and contributing to precision medicine.

One-step spectral clustering of weighted variables on single-cell RNA-sequencing data (단세포 RNA 시퀀싱 데이터를 위한 가중변수 스펙트럼 군집화 기법)

  • Park, Min Young;Park, Seyoung
    • The Korean Journal of Applied Statistics
    • /
    • v.33 no.4
    • /
    • pp.511-526
    • /
    • 2020
  • Single-cell RNA-sequencing (scRNA-seq) data consists of each cell's RNA expression extracted from large populations of cells. One main purpose of using scRNA-seq data is to identify inter-cellular heterogeneity. However, scRNA-seq data pose statistical challenges when applying traditional clustering methods because they have many missing values and high level of noise due to technical and sampling issues. In this paper, motivated by analyzing scRNA-seq data, we propose a novel spectral-based clustering method by imposing different weights on genes when computing a similarity between cells. Assigning weights on genes and clustering cells are performed simultaneously in the proposed clustering framework. We solve the proposed non-convex optimization using an iterative algorithm. Both real data application and simulation study suggest that the proposed clustering method better identifies underlying clusters compared with existing clustering methods.

Single-cell RNA-Seq unveils tumor microenvironment

  • Lee, Hae-Ock;Park, Woong-Yang
    • BMB Reports
    • /
    • v.50 no.6
    • /
    • pp.283-284
    • /
    • 2017
  • Single cell transcriptome analysis is a powerful tool for defining cell types or sub-populations within a heterogeneous bulk population. Tumor-associated microenvironment is a complex ecosystem consisting of numerous cell types that support tumor growth, angiogenesis, immune evasion, and metastasis. With the success of checkpoint inhibitors targeting the immune cell compartment, tumor microenvironment is emerging as a potential anti-cancer target, and understanding it has become an imminent subject in cancer biology.

A semi-automatic cell type annotation method for single-cell RNA sequencing dataset

  • Kim, Wan;Yoon, Sung Min;Kim, Sangsoo
    • Genomics & Informatics
    • /
    • v.18 no.3
    • /
    • pp.26.1-26.6
    • /
    • 2020
  • Single-cell RNA sequencing (scRNA-seq) has been widely applied to provide insights into the cell-by-cell expression difference in a given bulk sample. Accordingly, numerous analysis methods have been developed. As it involves simultaneous analyses of many cell and genes, efficiency of the methods is crucial. The conventional cell type annotation method is laborious and subjective. Here we propose a semi-automatic method that calculates a normalized score for each cell type based on user-supplied cell type-specific marker gene list. The method was applied to a publicly available scRNA-seq data of mouse cardiac non-myocyte cell pool. Annotating the 35 t-stochastic neighbor embedding clusters into 12 cell types was straightforward, and its accuracy was evaluated by constructing co-expression network for each cell type. Gene Ontology analysis was congruent with the annotated cell type and the corollary regulatory network analysis showed upstream transcription factors that have well supported literature evidences. The source code is available as an R script upon request.

The Workflow for Computational Analysis of Single-cell RNA-sequencing Data (단일 세포 RNA 시퀀싱 데이터에 대한 컴퓨터 분석의 작업과정)

  • Sung-Hun WOO;Byung Chul JUNG
    • Korean Journal of Clinical Laboratory Science
    • /
    • v.56 no.1
    • /
    • pp.10-20
    • /
    • 2024
  • RNA-sequencing (RNA-seq) is a technique used for providing global patterns of transcriptomes in samples. However, it can only provide the average gene expression across cells and does not address the heterogeneity within the samples. The advances in single-cell RNA sequencing (scRNA-seq) technology have revolutionized our understanding of heterogeneity and the dynamics of gene expression at the single-cell level. For example, scRNA-seq allows us to identify the cell types in complex tissues, which can provide information regarding the alteration of the cell population by perturbations, such as genetic modification. Since its initial introduction, scRNA-seq has rapidly become popular, leading to the development of a huge number of bioinformatic tools. However, the analysis of the big dataset generated from scRNA-seq requires a general understanding of the preprocessing of the dataset and a variety of analytical techniques. Here, we present an overview of the workflow involved in analyzing the scRNA-seq dataset. First, we describe the preprocessing of the dataset, including quality control, normalization, and dimensionality reduction. Then, we introduce the downstream analysis provided with the most commonly used computational packages. This review aims to provide a workflow guideline for new researchers interested in this field.

Single-Cell Toolkits Opening a New Era for Cell Engineering

  • Lee, Sean;Kim, Jireh;Park, Jong-Eun
    • Molecules and Cells
    • /
    • v.44 no.3
    • /
    • pp.127-135
    • /
    • 2021
  • Since the introduction of RNA sequencing (RNA-seq) as a high-throughput mRNA expression analysis tool, this procedure has been increasingly implemented to identify cell-level transcriptome changes in a myriad of model systems. However, early methods processed cell samples in bulk, and therefore the unique transcriptomic patterns of individual cells would be lost due to data averaging. Nonetheless, the recent and continuous development of new single-cell RNA sequencing (scRNA-seq) toolkits has enabled researchers to compare transcriptomes at a single-cell resolution, thus facilitating the analysis of individual cellular features and a deeper understanding of cellular functions. Nonetheless, the rapid evolution of high throughput single-cell "omics" tools has created the need for effective hypothesis verification strategies. Particularly, this issue could be addressed by coupling cell engineering techniques with single-cell sequencing. This approach has been successfully employed to gain further insights into disease pathogenesis and the dynamics of differentiation trajectories. Therefore, this review will discuss the current status of cell engineering toolkits and their contributions to single-cell and genome-wide data collection and analyses.