$\require{mediawiki-texvc}$

연합인증

연합인증 가입 기관의 연구자들은 소속기관의 인증정보(ID와 암호)를 이용해 다른 대학, 연구기관, 서비스 공급자의 다양한 온라인 자원과 연구 데이터를 이용할 수 있습니다.

이는 여행자가 자국에서 발행 받은 여권으로 세계 각국을 자유롭게 여행할 수 있는 것과 같습니다.

연합인증으로 이용이 가능한 서비스는 NTIS, DataON, Edison, Kafe, Webinar 등이 있습니다.

한번의 인증절차만으로 연합인증 가입 서비스에 추가 로그인 없이 이용이 가능합니다.

다만, 연합인증을 위해서는 최초 1회만 인증 절차가 필요합니다. (회원이 아닐 경우 회원 가입이 필요합니다.)

연합인증 절차는 다음과 같습니다.

최초이용시에는
ScienceON에 로그인 → 연합인증 서비스 접속 → 로그인 (본인 확인 또는 회원가입) → 서비스 이용

그 이후에는
ScienceON 로그인 → 연합인증 서비스 접속 → 서비스 이용

연합인증을 활용하시면 KISTI가 제공하는 다양한 서비스를 편리하게 이용하실 수 있습니다.

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

Korean journal of clinical laboratory science : KJCLS = 대한임상검사과학회지, v.56 no.1, 2024년, pp.10 - 20  

우성훈 (연세대학교 소프트웨어디지털헬스케어융합대학 임상병리학과) ,  정병출 (캘리포니아대학교 버클리캠퍼스 영양과학 및 독성학과)

초록
AI-Helper 아이콘AI-Helper

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.

Abstract AI-Helper 아이콘AI-Helper

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 ...

주제어

참고문헌 (58)

  1. Woo SH, Jung BC. Big data analytics in RNA-sequencing. Korean?J Clin Lab Sci. 2023;55:235-243. https://doi.org/10.15324/kjcls.2023.55.4.235 

  2. Jung BC, You D, Lee I, Li D, Schill RL, Ma K, et al. TET3 plays a?critical role in white adipose development and diet-induced?remodeling. Cell Rep. 2023;42:113196. https://doi.org/10.1016/j.celrep.2023.113196 

  3. Kim TK, Bae EJ, Jung BC, Choi M, Shin SJ, Park SJ, et al.?Inflammation promotes synucleinopathy propagation. Exp Mol?Med. 2022;54:2148-2161. https://doi.org/10.1038/s12276-022-00895-w 

  4. Park S, Lee C, Ku BM, Kim M, Park WY, Kim NKD, et al. Paired?analysis of tumor mutation burden calculated by targeted deep?sequencing panel and whole exome sequencing in non-small cell?lung cancer. BMB Rep. 2021;54:386-391. https://doi.org/10.5483/bmbrep.2021.54.7.045 

  5. Li X, Wang CY. From bulk, single-cell to spatial RNA sequencing.?Int J Oral Sci. 2021;13:36. https://doi.org/10.1038/s41368-021-00146-0 

  6. Tang F, Barbacioru C, Wang Y, Nordman E, Lee C, Xu N, et al.?mRNA-Seq whole-transcriptome analysis of a single cell. Nat?Methods. 2009;6:377-382. https://doi.org/10.1038/nmeth.1315 

  7. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018;50:1-14. https://doi.org/10.1038/s12276-018-0071-8 

  8. Haque A, Engel J, Teichmann SA, Lonnberg T. A practical guide?to single-cell RNA-sequencing for biomedical research and clinical applications. Genome Med. 2017;9:75. https://doi.org/10.1186/s13073-017-0467-4 

  9. Mair F, Erickson JR, Voillet V, Simoni Y, Bi T, Tyznik AJ, et al. A?targeted multi-omic analysis approach measures protein expression and low-abundance transcripts on the single-cell level.?Cell Rep. 2020;31:107499. https://doi.org/10.1016/j.celrep.2020.03.063 

  10. Zheng GX, Terry JM, Belgrader P, Ryvkin P, Bent ZW, Wilson R, et?al. Massively parallel digital transcriptional profiling of single cells.?Nat Commun. 2017;8:14049. https://doi.org/10.1038/ncomms14049 

  11. Lun ATL, Riesenfeld S, Andrews T, Dao TP, Gomes T, Marioni JC.?EmptyDrops: distinguishing cells from empty droplets in droplet-based single-cell RNA sequencing data. Genome Biol.?2019;20:63. https://doi.org/10.1186/s13059-019-1662-y 

  12. Yang S, Corbett SE, Koga Y, Wang Z, Johnson WE, Yajima M, et al.?Decontamination of ambient RNA in single-cell RNA-seq with?DecontX. Genome Biol. 2020;21:57. https://doi.org/10.1186/s13059-020-1950-6 

  13. Ilicic T, Kim JK, Kolodziejczyk AA, Bagger FO, McCarthy DJ,?Marioni JC, et al. Classification of low quality cells from single-cell RNA-seq data. Genome Biol. 2016;17:29. https://doi.org/10.1186/s13059-016-0888-1 

  14. Osorio D, Cai JJ. Systematic determination of the mitochondrial?proportion in human and mice tissues for single-cell RNA-sequencing data quality control. Bioinformatics. 2021;37:963-967.?https://doi.org/10.1093/bioinformatics/btaa751 

  15. Emont MP, Jacobs C, Essene AL, Pant D, Tenen D, Colleluori G, et?al. A single-cell atlas of human and mouse white adipose tissue.?Nature. 2022;603:926-933. https://doi.org/10.1038/s41586-022-04518-2 

  16. Wolock SL, Lopez R, Klein AM. Scrublet: computational identification of cell doublets in single-cell transcriptomic data. Cell?Syst. 2019;8:281-291.e9. https://doi.org/10.1016/j.cels.2018.11.005 

  17. McGinnis CS, Murrow LM, Gartner ZJ. DoubletFinder: doublet detection in single-cell RNA sequencing data using artificial nearest?neighbors. Cell Syst. 2019;8:329-337.e4. https://doi.org/10.1016/j.cels.2019.03.003 

  18. Xi NM, Li JJ. Benchmarking computational doublet-detection?methods for single-cell RNA sequencing data. Cell Syst. 2021;12:176-194.e6. https://doi.org/10.1016/j.cels.2020.11.008 

  19. Lu J, Sheng Y, Qian W, Pan M, Zhao X, Ge Q. scRNA-seq data analysis method to improve analysis performance. IET Nanobiotechnol.?2023;17:246-256. https://doi.org/10.1049/nbt2.12115 

  20. Wu Y, Zhang K. Tools for the analysis of high-dimensional single-cell RNA sequencing data. Nat Rev Nephrol. 2020;16:408-421.?https://doi.org/10.1038/s41581-020-0262-0 

  21. Lun AT, Bach K, Marioni JC. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome?Biol. 2016;17:75. https://doi.org/10.1186/s13059-016-0947-7 

  22. Vallejos CA, Risso D, Scialdone A, Dudoit S, Marioni JC.?Normalizing single-cell RNA sequencing data: challenges and?opportunities. Nat Methods. 2017;14:565-571. https://doi.org/10.1038/nmeth.4292 

  23. Choudhary S, Satija R. Comparison and evaluation of statistical?error models for scRNA-seq. Genome Biol. 2022;23:27. https://doi.org/10.1186/s13059-021-02584-9 

  24. Hafemeister C, Satija R. Normalization and variance stabilization?of single-cell RNA-seq data using regularized negative binomial?regression. Genome Biol. 2019;20:296. https://doi.org/10.1186/s13059-019-1874-1 

  25. Chandrashekar G, Sahin F. A survey on feature selection methods.?Comput Electr Eng. 2014;40:16-28. https://doi.org/10.1016/j.compeleceng.2013.11.024 

  26. Yang P, Huang H, Liu C. Feature selection revisited in the single-cell era. Genome Biol. 2021;22:321. https://doi.org/10.1186/s13059-021-02544-3 

  27. Sheng J, Li WV. Selecting gene features for unsupervised analysis?of single-cell gene expression data. Brief Bioinform. 2021;22:bbab295. https://doi.org/10.1093/bib/bbab295 

  28. Luecken MD, Theis FJ. Current best practices in single-cell?RNA-seq analysis: a tutorial. Mol Syst Biol. 2019;15:e8746.?https://doi.org/10.15252/msb.20188746 

  29. Do VH, Canzar S. A generalization of t-SNE and UMAP to single-cell multimodal omics. Genome Biol. 2021;22:130. https://doi.org/10.1186/s13059-021-02356-5 

  30. Meyer BH, Pozo ATR, Nunan Zola WM. Global and local structure?preserving GPU t-SNE methods for large-scale applications.?Expert Syst Appl. 2022;201:116918. https://doi.org/10.1016/j.eswa.2022.116918 

  31. Lee JA, Renard E, Bernard G, Dupont P, Verleysen M. Type 1 and 2?mixtures of Kullback-Leibler divergences as cost functions in dimensionality reduction based on similarity preservation. Neurocomputing. 2013;112:92-108. https://doi.org/10.1016/j.neucom.2012.12.036 

  32. Ge H, Zhu Z, Lou K, Wei W, Liu R, Damasevicius R, et al.?Classification of infrared objects in manifold space using Kullback-Leibler divergence of gaussian distributions of image points.?Symmetry. 2020;12:434. https://doi.org/10.3390/sym12030434 

  33. Kobak D, Berens P. The art of using t-SNE for single-cell?transcriptomics. Nat Commun. 2019;10:5416. https://doi.org/10.1038/s41467-019-13056-x 

  34. Becht E, McInnes L, Healy J, Dutertre CA, Kwok IWH, Ng LG, et al.?Dimensionality reduction for visualizing single-cell data using?UMAP. Nat Biotechnol. 2018. [Epub ahead of print]. https://doi.org/10.1038/nbt.4314 

  35. Sainburg T, McInnes L, Gentner TQ. Parametric UMAP embed-dings for representation and semisupervised learning. Neural?Comput. 2021;33:2881-2907. https://doi.org/10.1162/neco_a_01434 

  36. Andrews TS, Kiselev VY, McCarthy D, Hemberg M. Tutorial:?guidelines for the computational analysis of single-cell RNA sequencing data. Nat Protoc. 2021;16:1-9. https://doi.org/10.1038/s41596-020-00409-w 

  37. Kobak D, Linderman GC. Initialization is critical for preserving?global data structure in both t-SNE and UMAP. Nat Biotechnol.?2021;39:156-157. https://doi.org/10.1038/s41587-020-00809-z 

  38. Traag VA, Waltman L, van Eck NJ. From Louvain to Leiden: guaranteeing well-connected communities. Sci Rep. 2019;9:5233.?https://doi.org/10.1038/s41598-019-41695-z 

  39. Hairol Anuar SH, Abas ZA, Yunos NM, Mohd Zaki NH, Hashim?NA, Mokhtar MF, et al. Comparison between Louvain and Leiden?algorithm for network structure: a review. J Phys Conf Ser. 2021;2129:012028. https://doi.org/10.1088/1742-6596/2129/1/012028 

  40. El Bouchefry K, de Souza RS. Learning in big data: introduction?to machine learning. In: Skoda P, Adam F, editors. Knowledge?discovery in big data from astronomy and earth observation:?AstroGeoInformatics. Elsevier: 2020. p. 225-249. 

  41. Wolf FA, Angerer P, Theis FJ. SCANPY: large-scale single-cell?gene expression data analysis. Genome Biol. 2018;19:15. https://doi.org/10.1186/s13059-017-1382-0 

  42. Martini E, Kunderfranco P, Peano C, Carullo P, Cremonesi M,?Schorn T, et al. Single-cell sequencing of mouse heart immune?infiltrate in pressure overload-driven heart failure reveals extent?of immune activation. Circulation. 2019;140:2089-2107. https://doi.org/10.1161/circulationaha.119.041694 

  43. Qiu P. Embracing the dropouts in single-cell RNA-seq analysis.?Nat Commun. 2020;11:1169. https://doi.org/10.1038/s41467-020-14976-9 

  44. Squair JW, Gautier M, Kathe C, Anderson MA, James ND, Hutson?TH, et al. Confronting false discoveries in single-cell differential?expression. Nat Commun. 2021;12:5692. https://doi.org/10.1038/s41467-021-25960-2 

  45. Soneson C, Robinson MD. Bias, robustness and scalability in single-cell differential expression analysis. Nat Methods. 2018;15:?255-261. https://doi.org/10.1038/nmeth.4612 

  46. Rauch A, Mandrup S. Transcriptional networks controlling stromal cell differentiation. Nat Rev Mol Cell Biol. 2021;22:465-482.?https://doi.org/10.1038/s41580-021-00357-7 

  47. Bertoli C, Skotheim JM, de Bruin RA. Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol. 2013;14:518-528. https://doi.org/10.1038/nrm3629 

  48. Song D, Li JJ. PseudotimeDE: inference of differential gene expression along cell pseudotime with well-calibrated p-values?from single-cell RNA sequencing data. Genome Biol. 2021;22:124.?https://doi.org/10.1186/s13059-021-02341-y 

  49. Saelens W, Cannoodt R, Todorov H, Saeys Y. A comparison of?single-cell trajectory inference methods. Nat Biotechnol. 2019;?37:547-554. https://doi.org/10.1038/s41587-019-0071-9 

  50. Greenblatt MB, Ono N, Ayturk UM, Debnath S, Lalani S. The un-mixing problem: a guide to applying single-cell RNA sequencing?to bone. J Bone Miner Res. 2019;34:1207-1219. https://doi.org/10.1002/jbmr.3802 

  51. Trapnell C, Cacchiarelli D, Grimsby J, Pokharel P, Li S, Morse M,?et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol.?2014;32:381-386. https://doi.org/10.1038/nbt.2859 

  52. Zhang Y, Tran D, Nguyen T, Dascalu SM, Harris FC Jr. A robust?and accurate single-cell data trajectory inference method using?ensemble pseudotime. BMC Bioinformatics. 2023;24:55. https://doi.org/10.1186/s12859-023-05179-2 

  53. Van de Sande B, Lee JS, Mutasa-Gottgens E, Naughton B, Bacon?W, Manning J, et al. Applications of single-cell RNA sequencing?in drug discovery and development. Nat Rev Drug Discov.?2023;22:496-520. https://doi.org/10.1038/s41573-023-00688-4 

  54. Duncavage EJ, Bagg A, Hasserjian RP, DiNardo CD, Godley LA,?Iacobucci I, et al. Genomic profiling for clinical decision making?in myeloid neoplasms and acute leukemia. Blood. 2022;140:2228-2247. https://doi.org/10.1182/blood.2022015853 

  55. Kim N, Eum HH, Lee HO. Clinical perspectives of single-cell RNA?sequencing. Biomolecules. 2021;11:1161. https://doi.org/10.3390/biom11081161 

  56. Riess JW, Gandara DR, Frampton GM, Madison R, Peled N, Bufill?JA, et al. Diverse EGFR exon 20 insertions and co-occurring molecular alterations identified by comprehensive genomic profiling of NSCLC. J Thorac Oncol. 2018;13:1560-1568. https://doi.org/10.1016/j.jtho.2018.06.019 

  57. Vincent MD, Kuruvilla MS, Leighl NB, Kamel-Reid S. Biomarkers?that currently affect clinical practice: EGFR, ALK, MET, KRAS.?Curr Oncol. 2012;19(Suppl 1):S33-S44. https://doi.org/10.3747/co.19.1149 

  58. Qian J, Olbrecht S, Boeckx B, Vos H, Laoui D, Etlioglu E, et al. A?pan-cancer blueprint of the heterogeneous tumor microenvironment revealed by single-cell profiling. Cell Res. 2020;30:745-762.?https://doi.org/10.1038/s41422-020-0355-0 

관련 콘텐츠

오픈액세스(OA) 유형

GOLD

오픈액세스 학술지에 출판된 논문

이 논문과 함께 이용한 콘텐츠

저작권 관리 안내
섹션별 컨텐츠 바로가기

AI-Helper ※ AI-Helper는 오픈소스 모델을 사용합니다.

AI-Helper 아이콘
AI-Helper
안녕하세요, AI-Helper입니다. 좌측 "선택된 텍스트"에서 텍스트를 선택하여 요약, 번역, 용어설명을 실행하세요.
※ AI-Helper는 부적절한 답변을 할 수 있습니다.

선택된 텍스트

맨위로