J Pathol Transl Med.  2023 Jan;57(1):43-51. 10.4132/jptm.2022.12.12.

Single-cell and spatial sequencing application in pathology

Affiliations
  • 1Department of Microbiology, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 2Precision Medicine Research Center/Integrated Research Center for Genome Polymorphism, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 3Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 4Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
  • 5Cancer Evolution Research Center, College of Medicine, The Catholic University of Korea, Seoul, Korea

Abstract

Traditionally, diagnostic pathology uses histology representing structural alterations in a disease’s cells and tissues. In many cases, however, it is supplemented by other morphology-based methods such as immunohistochemistry and fluorescent in situ hybridization. Single-cell RNA sequencing (scRNA-seq) is one of the strategies that may help tackle the heterogeneous cells in a disease, but it does not usually provide histologic information. Spatial sequencing is designed to assign cell types, subtypes, or states according to the mRNA expression on a histological section by RNA sequencing. It can provide mRNA expressions not only of diseased cells, such as cancer cells but also of stromal cells, such as immune cells, fibroblasts, and vascular cells. In this review, we studied current methods of spatial transcriptome sequencing based on their technical backgrounds, tissue preparation, and analytic procedures. With the pathology examples, useful recommendations for pathologists who are just getting started to use spatial sequencing analysis in research are provided here. In addition, leveraging spatial sequencing by integration with scRNA-seq is reviewed. With the advantages of simultaneous histologic and single-cell information, spatial sequencing may give a molecular basis for pathological diagnosis, improve our understanding of diseases, and have potential clinical applications in prognostics and diagnostic pathology.

Keyword

Single-cell sequencing; Spatial sequencing; Pathology; Histology; Transcriptome; Diseases

Figure

  • Fig. 1 Overview of strengths and limitations of clinical and experimental methods for gene expression. Conventional methods of histopathology (A) and bulk RNA sequencing (RNA-seq) (B). FISH, fluorescence in situ hybridization. Recent methods of single-cell RNA sequencing (scRNA-seq) (C) and next-generation sequencing (NGS)-based spatial transcriptomics (ST) (D).

  • Fig. 2 Overview of spatial transcriptomics (ST) analysis. (A) Example of tissue slide (Visium technology) for ST. Original tissue image, detected area, spot clustering of ST data by unsupervised clustering (spatial spots colored by spot clusters), and magnified view of spatial spots are shown. Distance between spatial spots were 100-μm, and each spot has a diameter of 55-μm. (B) ST can measure genome-wide expression profiles in each 55-μm spatial spots. (C) Analysis strategies for ST data.

  • Fig. 3 Schematic view of integrated analysis of single-cell RNA sequencing (scRNA-seq) and spatial transcriptomics (ST). (A) Strengths and limitations of integrated analysis of scRNA-seq and ST. (B) Analysis strategies for integrated scRNA-seq and ST data.

  • Fig. 4 Example study of integrated analysis [38]. Findings from single-cell RNA sequencing (scRNA-seq) (A), spatial transcriptomics (ST) (B), and integrated analysis of scRNA-seq and ST (C) are summarized. FBR, foreign body reaction.


Reference

References

1. Stewart BJ, Clatworthy MR. Applying single-cell technologies to clinical pathology: progress in nephropathology. J Pathol. 2020; 250:693–704.
Article
2. Hwang B, Lee JH, Bang D. Single-cell RNA sequencing technologies and bioinformatics pipelines. Exp Mol Med. 2018; 50:1–14.
Article
3. Liu B, Li Y, Zhang L. Analysis and visualization of spatial transcriptomic data. Front Genet. 2021; 12:785290.
Article
4. Longo SK, Guo MG, Ji AL, Khavari PA. Integrating single-cell and spatial transcriptomics to elucidate intercellular tissue dynamics. Nat Rev Genet. 2021; 22:627–44.
Article
5. Tang F, Barbacioru C, Wang Y, et al. mRNA-Seq whole-transcriptome analysis of a single cell. Nat Methods. 2009; 6:377–82.
Article
6. Zheng GX, Terry JM, Belgrader P, et al. Massively parallel digital transcriptional profiling of single cells. Nat Commun. 2017; 8:14049.
Article
7. Chen G, Ning B, Shi T. Single-cell RNA-seq technologies and related computational data analysis. Front Genet. 2019; 10:317.
Article
8. Luecken MD, Theis FJ. Current best practices in single-cell RNA-seq analysis: a tutorial. Mol Syst Biol. 2019; 15:e8746.
Article
9. 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.
Article
10. Kashima Y, Sakamoto Y, Kaneko K, Seki M, Suzuki Y, Suzuki A. Single-cell sequencing techniques from individual to multiomics analyses. Exp Mol Med. 2020; 52:1419–27.
Article
11. Papalexi E, Satija R. Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol. 2018; 18:35–45.
Article
12. Ziegenhain C, Vieth B, Parekh S, et al. Comparative analysis of single-cell RNA sequencing methods. Mol Cell. 2017; 65:631–43.
Article
13. Rao A, Barkley D, Franca GS, Yanai I. Exploring tissue architecture using spatial transcriptomics. Nature. 2021; 596:211–20.
Article
14. Lewis SM, Asselin-Labat ML, Nguyen Q, et al. Spatial omics and multiplexed imaging to explore cancer biology. Nat Methods. 2021; 18:997–1012.
Article
15. Williams CG, Lee HJ, Asatsuma T, Vento-Tormo R, Haque A. An introduction to spatial transcriptomics for biomedical research. Genome Med. 2022; 14:68.
Article
16. Moor AE, Itzkovitz S. Spatial transcriptomics: paving the way for tissue-level systems biology. Curr Opin Biotechnol. 2017; 46:126–33.
Article
17. Jovic D, Liang X, Zeng H, Lin L, Xu F, Luo Y. Single-cell RNA sequencing technologies and applications: a brief overview. Clin Transl Med. 2022; 12:e694.
Article
18. 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.
Article
19. Denisenko E, Guo BB, Jones M, et al. Systematic assessment of tissue dissociation and storage biases in single-cell and single-nucleus RNA-seq workflows. Genome Biol. 2020; 21:130.
Article
20. Lafzi A, Moutinho C, Picelli S, Heyn H. Tutorial: guidelines for the experimental design of single-cell RNA sequencing studies. Nat Protoc. 2018; 13:2742–57.
Article
21. Slyper M, Porter CBM, Ashenberg O, et al. A single-cell and single-nucleus RNA-Seq toolbox for fresh and frozen human tumors. Nat Med. 2020; 26:792–802.
Article
22. Hao Y, Hao S, Andersen-Nissen E, et al. Integrated analysis of multimodal single-cell data. Cell. 2021; 184:3573–87.
Article
23. Aran D, Looney AP, Liu L, et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat Immunol. 2019; 20:163–72.
Article
24. Trapnell C, Cacchiarelli D, Grimsby J, et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat Biotechnol. 2014; 32:381–6.
Article
25. La Manno G, Soldatov R, Zeisel A, et al. RNA velocity of single cells. Nature. 2018; 560:494–8.
Article
26. Pratapa A, Jalihal AP, Law JN, Bharadwaj A, Murali TM. Benchmarking algorithms for gene regulatory network inference from single-cell transcriptomic data. Nat Methods. 2020; 17:147–54.
Article
27. Skok Gibbs C, Jackson CA, Saldi GA, et al. High-performance single-cell gene regulatory network inference at scale: the Inferelator 3.0. Bioinformatics. 2022; 38:2519–28.
Article
28. Zhuang X. Spatially resolved single-cell genomics and transcriptomics by imaging. Nat Methods. 2021; 18:18–22.
Article
29. Larsson L, Frisen J, Lundeberg J. Spatially resolved transcriptomics adds a new dimension to genomics. Nat Methods. 2021; 18:15–8.
Article
30. Villacampa EG, Larsson L, Mirzazadeh R, et al. Genome-wide spatial expression profiling in formalin-fixed tissues. Cell Genomics. 2021; 1:100065.
Article
31. Elosua-Bayes M, Nieto P, Mereu E, Gut I, Heyn H. SPOTlight: seeded NMF regression to deconvolute spatial transcriptomics spots with single-cell transcriptomes. Nucleic Acids Res. 2021; 49:e50.
Article
32. Bergenstrahle L, He B, Bergenstrahle J, et al. Super-resolved spatial transcriptomics by deep data fusion. Nat Biotechnol. 2022; 40:476–9.
Article
33. Qian X, Harris KD, Hauling T, et al. Probabilistic cell typing enables fine mapping of closely related cell types in situ. Nat Methods. 2020; 17:101–6.
Article
34. Arora R, Cao C, Kumar M, et al. Spatial transcriptomics reveals distinct and conserved tumor core and edge architectures that predict survival and targeted therapy response. Preprint at: https://doi.org/10.1101/2022.09.04.505581 . 2022.
Article
35. Armingol E, Officer A, Harismendy O, Lewis NE. Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet. 2021; 22:71–88.
Article
36. Efremova M, Vento-Tormo M, Teichmann SA, Vento-Tormo R. CellPhoneDB: inferring cell-cell communication from combined expression of multi-subunit ligand-receptor complexes. Nat Protoc. 2020; 15:1484–506.
Article
37. Dries R, Zhu Q, Dong R, et al. Giotto: a toolbox for integrative analysis and visualization of spatial expression data. Genome Biol. 2021; 22:78.
Article
38. Kim YS, Shin S, Choi EJ, et al. Different molecular features of epithelioid and giant cells in foreign body reaction identified by single-cell RNA sequencing. J Invest Dermatol. 2022; 142:3232–42.
Article
39. Ji AL, Rubin AJ, Thrane K, et al. Multimodal analysis of composition and spatial architecture in human squamous cell carcinoma. Cell. 2020; 182:497–514.
Article
40. Moncada R, Barkley D, Wagner F, et al. Integrating microarray-based spatial transcriptomics and single-cell RNA-seq reveals tissue architecture in pancreatic ductal adenocarcinomas. Nat Biotechnol. 2020; 38:333–42.
Article
41. Argelaguet R, Clark SJ, Mohammed H, et al. Multi-omics profiling of mouse gastrulation at single-cell resolution. Nature. 2019; 576:487–91.
Article
42. Abascal F, Harvey LM, Mitchell E, et al. Somatic mutation landscapes at single-molecule resolution. Nature. 2021; 593:405–10.
Article
43. Lee J, Hyeon DY, Hwang D. Single-cell multiomics: technologies and data analysis methods. Exp Mol Med. 2020; 52:1428–42.
Article
Full Text Links
  • JPTM
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr