• 제목/요약/키워드: RNA sequencing (RNA-seq)

검색결과 155건 처리시간 0.026초

RNA-Seq 정렬 알고리즘의 동향 (Recent Trends in RNA-Seq Alignment Algorithms)

  • 유승학;최민석;윤성로
    • 한국정보처리학회:학술대회논문집
    • /
    • 한국정보처리학회 2014년도 추계학술발표대회
    • /
    • pp.669-671
    • /
    • 2014
  • High Throughput Sequencing (HTS) 기술의 발달로 인해 시퀀싱 비용이 감소함에 따라 다양한 분야에서 이를 활용한 융합 연구가 활발하게 진행되고 있다. HTS 기술에서 가장 중요한 부분은 수백만개의 short read 들을 표준유전체 (reference genome)에 정렬시키는 것인데 RNA 시퀀싱 (RNA-Seq) 의 경우 RNA splicing 으로 인해 일반적인 aligner 로 처리가 불가능하다. 복잡한 RNA-Seq 정렬 문제를 해결하기 위해 그동안 다양한 알고리즘들이 제안되어 왔다. 본 논문에서는 RNA-seq 정렬분야에서 잘 알려진 알고리즘들과 최신 알고리즘들을 살펴봄으로써 RNA-seq 정렬 알고리즘의 동향을 살펴보고자 한다.

TRAPR: R Package for Statistical Analysis and Visualization of RNA-Seq Data

  • Lim, Jae Hyun;Lee, Soo Youn;Kim, Ju Han
    • Genomics & Informatics
    • /
    • 제15권1호
    • /
    • pp.51-53
    • /
    • 2017
  • High-throughput transcriptome sequencing, also known as RNA sequencing (RNA-Seq), is a standard technology for measuring gene expression with unprecedented accuracy. Numerous bioconductor packages have been developed for the statistical analysis of RNA-Seq data. However, these tools focus on specific aspects of the data analysis pipeline, and are difficult to appropriately integrate with one another due to their disparate data structures and processing methods. They also lack visualization methods to confirm the integrity of the data and the process. In this paper, we propose an R-based RNA-Seq analysis pipeline called TRAPR, an integrated tool that facilitates the statistical analysis and visualization of RNA-Seq expression data. TRAPR provides various functions for data management, the filtering of low-quality data, normalization, transformation, statistical analysis, data visualization, and result visualization that allow researchers to build customized analysis pipelines.

Strategy of Patient-Specific Therapeutics in Cardiovascular Disease Through Single-Cell RNA Sequencing

  • Yunseo Jung;Juyeong Kim;Howon Jang;Gwanhyeon Kim;Yoo-Wook Kwon
    • Korean Circulation Journal
    • /
    • 제53권1호
    • /
    • pp.1-16
    • /
    • 2023
  • Recently, single cell RNA sequencing (scRNA-seq) technology has enabled the discovery of novel or rare subtypes of cells and their characteristics. This technique has advanced unprecedented biomedical research by enabling the profiling and analysis of the transcriptomes of single cells at high resolution and throughput. Thus, scRNA-seq has contributed to recent advances in cardiovascular research by the generation of cell atlases of heart and blood vessels and the elucidation of mechanisms involved in cardiovascular development and diseases. This review summarizes the overall workflow of the scRNA-seq technique itself and key findings in the cardiovascular development and diseases based on the previous studies. In particular, we focused on how the single-cell sequencing technology can be utilized in clinical field and precision medicine to treat specific diseases.

Use of cutting-edge RNA-sequencing technology to identify biomarkers and potential therapeutic targets in canine and feline cancers and other diseases

  • Youngdong Choi;Min-Woo Nam;Hong Kyu Lee;Kyung-Chul Choi
    • Journal of Veterinary Science
    • /
    • 제24권5호
    • /
    • pp.71.1-71.12
    • /
    • 2023
  • With the growing interest in companion animals and the rapidly expanding animal healthcare and pharmaceuticals market worldwide. With the advancements in RNAsequencing (RNA-seq) technology, it has become a valuable tool for understanding biological processes in companion animals and has multiple applications in animal healthcare. Historically, veterinary diagnoses and treatments relied solely on clinical symptoms and drugs used in human diseases. However, RNA-seq has emerged as an effective technology for studying companion animals, providing insights into their genetic information. The sequencing technology has revealed that not only messenger RNAs (mRNAs) but also noncoding RNAs (ncRNAs) such as long ncRNAs and microRNAs can serve as biomarkers. Based on the examination of RNA-seq applications in veterinary medicine, particularly in dogs and cats, this review concludes that RNA-seq has significant potential as a diagnostic and research tool. It has enabled the identification of potential biomarkers for cancer and other diseases in companion animals. Further research and development are required to maximize the utilization of RNA-seq for improved disease diagnosis and therapeutic targeting in companion animals.

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

  • 우성훈;정병출
    • 대한임상검사과학회지
    • /
    • 제56권1호
    • /
    • pp.10-20
    • /
    • 2024
  • RNA-시퀀싱은 표본에 대한 전사체 전체의 패턴을 제공하는 기법이다. 그러나 RNA-시퀀싱은 표본 내 전체 세포에 대한 평균 유전자 발현만 제공할 수 있으며, 표본 내의 이질성(heterogeneity)에 대한 정보는 제공하지 못한다. 단일 세포 RNA-시퀀싱 기술의 발전을 통해 우리는 표본의 단일 세포 수준에서 이질성과 유전자 발현의 동역학(dynamics)에 대한 이해를 할 수 있게 되었다. 예를 들어, 우리는 단일 세포 RNA-시퀀싱을 통해 복잡한 조직을 구성하는 다양한 세포 유형을 식별할 수 있으며, 특정 세포 유형의 유전자 발현 변화와 같은 정보를 알 수 있다. 단일 세포 RNA-시퀀싱은 처음 도입된 이후 많은 이들의 관심을 끌게 되었으며, 이를 활용하기 위한 대규모 생물정보학(bioinformatics) 도구가 개발되었다. 그러나 단일 세포 RNA-시퀀싱에서 생성된 빅데이터 분석에는 데이터 전처리에 대한 이해와 전처리 이후 다양한 분석 기술에 대한 이해가 필요하다. 본 종설에서는 단일 세포 RNA-시퀀싱 데이터분석과 관련된 작업과정의 개요를 제시한다. 먼저 데이터의 품질 관리, 정규화 및 차원 감소와 같은 데이터의 전 처리 과정에 대해 설명한다. 그 이후, 가장 일반적으로 사용되는 생물정보학 도구를 활용한 데이터의 후속 분석에 대해 설명한다. 본 종설은 이 분야에 관심이 있는 새로운 연구자를 위한 가이드라인을 제공하는 것을 목표로 한다.

앙상블 기법을 활용한 RNA-Sequencing 데이터의 폐암 예측 연구 (A Study on Predicting Lung Cancer Using RNA-Sequencing Data with Ensemble Learning)

  • Geon AN;JooYong PARK
    • Journal of Korea Artificial Intelligence Association
    • /
    • 제2권1호
    • /
    • pp.7-14
    • /
    • 2024
  • In this paper, we explore the application of RNA-sequencing data and ensemble machine learning to predict lung cancer and treatment strategies for lung cancer, a leading cause of cancer mortality worldwide. The research utilizes Random Forest, XGBoost, and LightGBM models to analyze gene expression profiles from extensive datasets, aiming to enhance predictive accuracy for lung cancer prognosis. The methodology focuses on preprocessing RNA-seq data to standardize expression levels across samples and applying ensemble algorithms to maximize prediction stability and reduce model overfitting. Key findings indicate that ensemble models, especially XGBoost, substantially outperform traditional predictive models. Significant genetic markers such as ADGRF5 is identified as crucial for predicting lung cancer outcomes. In conclusion, ensemble learning using RNA-seq data proves highly effective in predicting lung cancer, suggesting a potential shift towards more precise and personalized treatment approaches. The results advocate for further integration of molecular and clinical data to refine diagnostic models and improve clinical outcomes, underscoring the critical role of advanced molecular diagnostics in enhancing patient survival rates and quality of life. This study lays the groundwork for future research in the application of RNA-sequencing data and ensemble machine learning techniques in clinical settings.

RNA-Seq 데이터를 이용한 선택 스플라이싱 유형 분석 (Alternative Splicing Pattern Analysis from RNA-Seq data)

  • 공진화;이종근;이은주;윤지희
    • 한국정보과학회:학술대회논문집
    • /
    • 한국정보과학회 2011년도 한국컴퓨터종합학술대회논문집 Vol.38 No.1(A)
    • /
    • pp.37-40
    • /
    • 2011
  • 선택 스플라이싱 (alternative splicing)은 mRNA (messenger RNA)의 전구체인 pre-mRNA가 mRNA로 전사될 때 pre-mRNA의 엑손 영역들 (exons)이 여러 가지 유형 (pattern)으로 다시 연결되는 과정을 말한다. 선택 스플라이싱에 의해 하나의 유전자로부터 서로 다른 mRNA가 만들어 지고 서로 다른 이소형의 단백질 (protein isoforms)이 생성된다. 현재까지 알려진 선택 스플라이싱의 유형은 약 7가지 종류가 있으며, 유전자의 돌연변이 및 질병과 밀접한 연관성을 가지고 있는 것으로 알려져 있다. 본 연구에서는 차세대 시퀀싱 (Next Generation Sequencing : NGS) 기술로 생성된 RNA-Seq 데이터로부터 각 유전자 영역에 대한 선택 스플라이싱 유형을 분류/추출하는 새로운 알고리즘을 제안한다. 제안된 알고리즘에서는 RNA-Seq 데이터를 DNA 시퀀스와 mRNA 트랜스크립트 시퀀스에 동시 매핑하고, 각 엑손 영역에 정렬된 RNA-Seq 데이터의 커버리지 정보 및 엑손의 접합 (junction) 정보를 이용하여 발현된 트랜스크립트 (transcript)의 종류와 양을 측정한다. 알고리즘의 유효성을 보이기 위하여 시뮬레이션 데이터를 이용한 인간 유전자 영역에서의 선택 스플라이싱 유형 추출 실험을 수행하였으며, 검증된 선택 스플라이싱 DB와 비교, 검증하였다.

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

  • Roth, Robert;Kim, Soochi;Kim, Jeesu;Rhee, Siyeon
    • BMB Reports
    • /
    • 제53권8호
    • /
    • 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.

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

  • Kim, Wan;Yoon, Sung Min;Kim, Sangsoo
    • Genomics & Informatics
    • /
    • 제18권3호
    • /
    • 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.

Transcriptional Heterogeneity of Cellular Senescence in Cancer

  • Junaid, Muhammad;Lee, Aejin;Kim, Jaehyung;Park, Tae Jun;Lim, Su Bin
    • Molecules and Cells
    • /
    • 제45권9호
    • /
    • pp.610-619
    • /
    • 2022
  • Cellular senescence plays a paradoxical role in tumorigenesis through the expression of diverse senescence-associated (SA) secretory phenotypes (SASPs). The heterogeneity of SA gene expression in cancer cells not only promotes cancer stemness but also protects these cells from chemotherapy. Despite the potential correlation between cancer and SA biomarkers, many transcriptional changes across distinct cell populations remain largely unknown. During the past decade, single-cell RNA sequencing (scRNA-seq) technologies have emerged as powerful experimental and analytical tools to dissect such diverse senescence-derived transcriptional changes. Here, we review the recent sequencing efforts that successfully characterized scRNA-seq data obtained from diverse cancer cells and elucidated the role of senescent cells in tumor malignancy. We further highlight the functional implications of SA genes expressed specifically in cancer and stromal cell populations in the tumor microenvironment. Translational research leveraging scRNA-seq profiling of SA genes will facilitate the identification of novel expression patterns underlying cancer susceptibility, providing new therapeutic opportunities in the era of precision medicine.