Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging

Subjects

Abstract

Resolving the spatial distribution of RNA and protein in tissues at subcellular resolution is a challenge in the field of spatial biology. We describe spatial molecular imaging, a system that measures RNAs and proteins in intact biological samples at subcellular resolution by performing multiple cycles of nucleic acid hybridization of fluorescent molecular barcodes. We demonstrate that spatial molecular imaging has high sensitivity (one or two copies per cell) and very low error rate (0.0092 false calls per cell) and background (~0.04 counts per cell). The imaging system generates three-dimensional, super-resolution localization of analytes at ~2 million cells per sample. Cell segmentation is morphology based using antibodies, compatible with formalin-fixed, paraffin-embedded samples. We measured multiomic data (980 RNAs and 108 proteins) at subcellular resolution in formalin-fixed, paraffin-embedded tissues (nonsmall cell lung and breast cancer) and identified >18 distinct cell types, ten unique tumor microenvironments and 100 pairwise ligand–receptor interactions. Data on >800,000 single cells and ~260 million transcripts can be accessed at http://nanostring.com/CosMx-dataset.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: CosMx SMI chemistry and workflow and 3D mapping of RNA at single-cell and subcellular resolution using morphology-based cell segmentation.
Fig. 2: Single-cell distribution and detection sensitivity of low, medium and high expressers and comparison of SMI data with RNA-seq and RNAscope data.
Fig. 3: Spatial RNA detection to identify cell types and cell–cell interactions in FFPE human NSCLC tissue.
Fig. 4: Paired LR expression between interacting tumor cells and T cells varies across tumors.
Fig. 5: Concordance between serial FFPE lung sections over a spatial grid.
Fig. 6: Spatial subcellular protein analysis on SMI.

Similar content being viewed by others

Data availability

The full RNA NSCLC dataset used in this study is available at http://nanostring.com/CosMx-dataset.

Code availability

Data from this publication (the full RNA NSCLC dataset) has been placed in the public domain in a format that can be analyzed and visualized using a variety of open-source packages, such as Seurat (https://github.com/satijalab/seurat) and Giotto (https://github.com/RubD/Giotto). Nearly all of the analyses performed in this paper can be accomplished using these open-source packages. For any of the specialized code/analyses performed in this manuscript that are not available through open-source packages, requests can be made through email to the corresponding author.

References

  1. Garon, E. B. et al. Pembrolizumab for the treatment of non-small-cell lung cancer. N. Engl. J. Med. 372, 2018–28 (2015).

    Article  Google Scholar 

  2. Yu, H. et al. PD-L1 expression by two complementary diagnostic assays and mRNA in situ hybridization in small cell lung cancer. J. Thorac. Oncol. 12, 110–120 (2017).

    Article  Google Scholar 

  3. Yu, H., Boyle, T. A., Zhou, C., Rimm, D. L. & Hirsch, F. R. PD-L1 expression in lung cancer. J .Thorac. Oncol. 11, 964–975 (2016).

    Article  Google Scholar 

  4. Ting, D. T. et al. Aberrant overexpression of satellite repeats in pancreatic and other epithelial cancers. Science 331, 593–596 (2011).

    Article  CAS  Google Scholar 

  5. Garber, K. Oncologists await historic first: a pan-tumor predictive marker, for immunotherapy. Nat. Biotechnol. 35, 297–298 (2017).

    Article  CAS  Google Scholar 

  6. Sokolenko, A. P. & Imyanitov, E. N. Molecular tests for the choice of cancer therapy. Curr. Pharm. Des. 23, 4794–4806 (2017).

    Article  CAS  Google Scholar 

  7. Dereli, A. S., Bailey, E. J. & Kumar, N. N. Combining multiplex fluorescence in situ hybridization with fluorescent immunohistochemistry on fresh frozen or fixed mouse brain sections. J. Vis. Exp. https://doi.org/10.3791/6170910.3791/61709 (2021).

  8. Taube, J. M. et al. The Society for Immunotherapy of Cancer statement on best practices for multiplex immunohistochemistry (IHC) and immunofluorescence (IF) staining and validation. J. Immunother. Cancer 8, e000155 (2020).

  9. Hirsch, F. R. et al. PD-L1 immunohistochemistry assays for lung cancer: results from Phase 1 of the Blueprint PD-L1 IHC Assay Comparison Project. J. Thorac. Oncol. 12, 208–222 (2017).

    Article  Google Scholar 

  10. Udall, M. et al. PD-L1 diagnostic tests: a systematic literature review of scoring algorithms and test-validation metrics. Diagn. Pathol. 13, 12 (2018).

    Article  Google Scholar 

  11. Halse, H. et al. Multiplex immunohistochemistry accurately defines the immune context of metastatic melanoma. Sci. Rep. 8, 11158 (2018).

    Article  CAS  Google Scholar 

  12. Macosko Evan, Z. et al. Highly parallel genome-wide expression profiling of individual cells using nanoliter droplets. Cell. 161, 1202–1214 (2015).

    Article  CAS  Google Scholar 

  13. Zilionis, R. et al. Single-cell barcoding and sequencing using droplet microfluidics. Nat. Protoc. 12, 44–73 (2017).

    Article  CAS  Google Scholar 

  14. Wang, X., He, Y., Zhang, Q., Ren, X. & Zhang, Z. Direct comparative analyses of 10X Genomics Chromium and Smart-seq2. Genomics Proteomics Bioinformatics 19, 253–266 (2021).

  15. See, P., Lum, J., Chen, J. & Ginhoux, F. A single-cell sequencing guide for immunologists. Front. Immunol. 9, 2425 (2018).

    Article  Google Scholar 

  16. Ståhl, P. L. et al. Visualization and analysis of gene expression in tissue sections by spatial transcriptomics. Science 353, 78–82 (2016).

    Article  Google Scholar 

  17. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2021).

    Article  CAS  Google Scholar 

  18. Fu, X. et al. Continuous polony gels for tissue mapping with high resolution and RNA capture efficiency. Preprint at bioRxiv https://doi.org/10.1101/2021.03.17.435795 (2021).

  19. Liu, Y. et al. High-spatial-resolution multi-omics sequencing via deterministic barcoding in tissue. Cell 183, 1665–1681 (2020).

    Article  CAS  Google Scholar 

  20. Merritt, C. R. et al. Multiplex digital spatial profiling of proteins and RNA in fixed tissue. Nat. Biotechnol. 38, 586–599 (2020).

    Article  CAS  Google Scholar 

  21. Wu, Q. et al. Poly A – transcripts expressed in HeLa cells. PLoS ONE 3, e2803 (2008).

    Article  Google Scholar 

  22. Moffitt, J. R. et al. High-throughput single-cell gene-expression profiling with multiplexed error-robust fluorescence in situ hybridization. Proc. Natl Acad. Sci. USA 113, 11046–11051 (2016).

    Article  CAS  Google Scholar 

  23. Moffitt, J. R. & Zhuang, X. in Methods in Enzymology (eds Filonov, G. S. & Jaffrey, S. R.) 1–49 (Academic Press, 2016).

  24. Groiss, S. et al. Highly resolved spatial transcriptomics for detection of rare events in cells. Preprint at bioRxiv https://doi.org/10.1101/2021.10.11.463936 (2021).

  25. Lee, J. H. et al. Fluorescent in situ sequencing (FISSEQ) of RNA for gene expression profiling in intact cells and tissues. Nat. Protoc. 10, 442–458 (2015).

    Article  CAS  Google Scholar 

  26. Eng, C.-H. L. et al. Transcriptome-scale super-resolved imaging in tissues by RNA seqFISH+. Nature 568, 235–239 (2019).

    Article  CAS  Google Scholar 

  27. Xia, C., Fan, J., Emanuel, G., Hao, J. & Zhuang, X. Spatial transcriptome profiling by MERFISH reveals subcellular RNA compartmentalization and cell cycle-dependent gene expression. Proc. Natl Acad. Sci. USA 116, 19490–19499 (2019).

    Article  CAS  Google Scholar 

  28. Goltsev, Y. et al. Deep profiling of mouse splenic architecture with CODEX multiplexed imaging. Cell 174, 968–981 (2018).

    Article  CAS  Google Scholar 

  29. Lin, J. R., Fallahi-Sichani, M. & Sorger, P. K. Highly multiplexed imaging of single cells using a high-throughput cyclic immunofluorescence method. Nat. Commun. 6, 8390 (2015).

    Article  CAS  Google Scholar 

  30. Giesen, C. et al. Highly multiplexed imaging of tumor tissues with subcellular resolution by mass cytometry. Nat. Methods 11, 417–422 (2014).

    Article  CAS  Google Scholar 

  31. Chen, K. H., Boettiger, A. N., Moffitt, J. R., Wang, S. & Zhuang, X. RNA imaging. Spatially resolved, highly multiplexed RNA profiling in single cells. Science 348, aaa6090 (2015).

    Article  Google Scholar 

  32. Mercer, T. R. et al. The human mitochondrial transcriptome. Cell 146, 645–658 (2011).

    Article  CAS  Google Scholar 

  33. Gudenas, B. L. & Wang, L. Prediction of LncRNA subcellular localization with deep learning from sequence features. Sci. Rep. 8, 16385 (2018).

    Article  Google Scholar 

  34. Baker, S. C. et al. The External RNA Controls Consortium: a progress report. Nat. Methods 2, 731–734 (2005).

    Article  CAS  Google Scholar 

  35. Ghandi, M. et al. Next-generation characterization of the Cancer Cell Line Encyclopedia. Nature 569, 503–508 (2019).

    Article  CAS  Google Scholar 

  36. Nusinow, D. P. et al. Quantitative proteomics of the Cancer Cell Line Encyclopedia. Cell 180, 387–402 (2020).

    Article  CAS  Google Scholar 

  37. National Cancer Institute. NCI-60 Human Tumor Cell Lines Screen https://dtp.cancer.gov/discovery_development/nci-60/ (2020).

  38. Kinker, G. S. et al. Pan-cancer single-cell RNA-seq identifies recurring programs of cellular heterogeneity. Nat. Genet. 52, 1208–1218 (2020).

    Article  CAS  Google Scholar 

  39. Liu, J. et al. Concordance of MERFISH spatial transcriptomics with bulk and single-cell RNA sequencing. Preprint at bioRxiv https://doi.org/10.1101/2022.03.04.483068 (2022).

  40. Leader, A. M. et al. Single-cell analysis of human non-small cell lung cancer lesions refines tumor classification and patient stratification. Cancer Cell 39, 1594–1609 (2021).

    Article  CAS  Google Scholar 

  41. Danaher, P. et al. Advances in mixed cell deconvolution enable quantification of cell types in spatially-resolved gene expression data. Nat. Commun. 13, 385 (2022).

  42. Illumina. Evaluating RNA quality from FFPE samples. https://www.illumina.com/content/dam/illumina-marketing/documents/products/technotes/evaluating-rna-quality-from-ffpe-samples-technical-note-470-2014-001.pdf (2021).

  43. Leica Biosystems. OND-III fully automated IHC and ISH staining system. https://www.leicabiosystems.com/ihc-ish-fish/fully-automated-ihc-ish-instruments/bond-iii/ (2021).

  44. Qiu, P. Embracing the dropouts in single-cell RNA-seq analysis. Nat. Commun. 11, 1169 (2020).

    Article  CAS  Google Scholar 

  45. Lin, J. R., Fallahi-Sichani, M., Chen, J. Y. & Sorger, P. K. Cyclic immunofluorescence (CycIF), a highly multiplexed method for single-cell imaging. Curr. Protoc. Chem. Biol. 8, 251–264 (2016).

    Article  Google Scholar 

  46. Caruthers, M. H. et al. Chemical synthesis of deoxyoligonucleotides by the phosphoramidite method. Methods Enzymol. 154, 287–313 (1987).

    Article  CAS  Google Scholar 

  47. Zhang, Z., Revyakin, A., Grimm, J. B., Lavis, L. D. & Tjian, R. Single-molecule tracking of the transcription cycle by sub-second RNA detection. eLife 3, e01775 (2014).

    Article  Google Scholar 

  48. Wagle, M.-C. et al. A transcriptional MAPK Pathway Activity Score (MPAS) is a clinically relevant biomarker in multiple cancer types. NPJ Precis. Oncol. 2, 7 (2018).

    Article  Google Scholar 

  49. Son, Y. H. et al. Roles of MAPK and NF-kappaB in interleukin-6 induction by lipopolysaccharide in vascular smooth muscle cells. J. Cardiovasc. Pharmacol. 51, 71–77 (2008).

    Article  CAS  Google Scholar 

  50. Kang, H. B., Kim, Y. E., Kwon, H. J., Sok, D. E. & Lee, Y. Enhancement of NF-kappaB expression and activity upon differentiation of human embryonic stem cell line SNUhES3. Stem Cells Dev. 16, 615–623 (2007).

    Article  CAS  Google Scholar 

  51. Hiscott, J. et al. Induction of human interferon gene expression is associated with a nuclear factor that interacts with the NF-kappa B site of the human immunodeficiency virus enhancer. J. Virol. 63, 2557–2566 (1989).

    Article  CAS  Google Scholar 

  52. Kitamura, A., Takahashi, K., Okajima, A. & Kitamura, N. Induction of the human gene for p44, a hepatitis-C-associated microtubular aggregate protein, by interferon-alpha/beta. Eur. J. Biochem. 224, 877–883 (1994).

    Article  CAS  Google Scholar 

  53. Kim, J. H., Park, S. Y., Jun, Y., Kim, J. Y. & Nam, J. S. Roles of Wnt target genes in the journey of cancer stem cells. Int. J. Mol. Sci. 18, 1604 (2017).

  54. Jho, E. H. et al. Wnt/beta-catenin/Tcf signaling induces the transcription of Axin2, a negative regulator of the signaling pathway. Mol. Cell. Biol. 22, 1172–1183 (2002).

    Article  CAS  Google Scholar 

  55. Lustig, B. et al. Negative feedback loop of Wnt signaling through upregulation of conductin/axin2 in colorectal and liver tumors. Mol. Cell. Biol. 22, 1184–1193 (2002).

    Article  CAS  Google Scholar 

  56. Yan, D. et al. Elevated expression of axin2 and hnkd mRNA provides evidence that Wnt/beta–catenin signaling is activated in human colon tumors. Proc. Natl Acad. Sci. USA 98, 14973–14978 (2001).

    Article  CAS  Google Scholar 

  57. Ramakrishnan, A. B. & Cadigan, K. M. Wnt target genes and where to find them. F1000Res 6, 746 (2017).

    Article  Google Scholar 

  58. Barker, N. et al. Identification of stem cells in small intestine and colon by marker gene Lgr5. Nature 449, 1003–1007 (2007).

    Article  CAS  Google Scholar 

  59. He, T. C. et al. Identification of c-MYC as a target of the APC pathway. Science 281, 1509–1512 (1998).

    Article  CAS  Google Scholar 

  60. Shtutman, M. et al. The cyclin D1 gene is a target of the beta-catenin/LEF-1 pathway. Proc. Natl Acad. Sci. USA 96, 5522–5527 (1999).

    Article  CAS  Google Scholar 

  61. Katoh, Y. & Katoh, M. Hedgehog target genes: mechanisms of carcinogenesis induced by aberrant hedgehog signaling activation. Curr. Mol. Med. 9, 873–886 (2009).

    Article  CAS  Google Scholar 

  62. Danaher, P. et al. Gene expression markers of Tumor Infiltrating Leukocytes. J. Immunother. Cancer 5, 18 (2017).

    Article  Google Scholar 

  63. Nguyen, Q. H. et al. Profiling human breast epithelial cells using single cell RNA sequencing identifies cell diversity. Nat. Commun. 9, 2028 (2018).

    Article  Google Scholar 

  64. Barneda, D. et al. The brown adipocyte protein CIDEA promotes lipid droplet fusion via a phosphatidic acid-binding amphipathic helix. eLife 4, e07485 (2015).

    Article  Google Scholar 

  65. Ussar, S. et al. ASC-1, PAT2, and P2RX5 are cell surface markers for white, beige, and brown adipocytes. Sci. Transl. Med. 6, 247ra103 (2014).

    Article  Google Scholar 

  66. Min, S. Y. et al. Diverse repertoire of human adipocyte subtypes develops from transcriptionally distinct mesenchymal progenitor cells. Proc. Natl Acad. Sci. USA 116, 17970–17979 (2019).

    Article  CAS  Google Scholar 

  67. Shan, T., Liu, W. & Kuang, S. Fatty acid binding protein 4 expression marks a population of adipocyte progenitors in white and brown adipose tissues. FASEB J. 27, 277–287 (2013).

    Article  CAS  Google Scholar 

  68. Bradley, D. & Roth, G. Adaptive thresholding using the integral image. J. Graph. Tools 12, 13–21 (2007).

    Article  Google Scholar 

  69. Krishnamoorthy, A. & Menon, D. Matrix inversion using Cholesky decomposition. In 2013 Signal Processing: Algorithms, Architectures, Arrangements, and Applications (SPA) (IEEE, 2013).

  70. Stringer, C., Wang, T., Michaelos, M. & Pachitariu, M. Cellpose: a generalist algorithm for cellular segmentation. Nat. Methods 18, 100–106 (2021).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

Research and development reported in this publication was supported in part through a strategic development collaboration between NanoString Technologies and Lam Research (Fremont, CA). The authors thank B. Birditt, B. Filanoski, J. Jenkins and E. Zhao from NanoString Technologies for provision of technical support.

Author information

Authors and Affiliations

Authors

Contributions

S.H. undertook conception and design of the work, supervised data collection, analysis and interpretation and drafted the manuscript. R.B. developed protein data processing algorithms. C.B. was responsible for instrument control software, SMI automation and integration of all instrument subsystems software. E.A.B. reviewed and validated protein data (Fig. 6) and performed the nested protein multiplex validation experiment (Supplementary Fig. 17). D.L.B. developed manual and automated processes for inventorying, quantitation, normalization, pooling, purification and quality control of oligonucleotides in 980-plex RNA panel and SMI reporters. K.C. performed oligo conjugations of antibodies used in SMI protein assays. P.D. designed the 980-plex RNA panel with E.P., performed comparison with RNA-seq in Fig. 2, performed NSCLC analyses in Fig. 3, performed reproducibility analysis in Fig. 5 and contributed to manuscript writing/editing. D.D. led the team that developed the system, instrumentation and software. R.G.G. developed reporter chemistry, assembly, quality control and manufacturing, and contributed to the section SMI reporter design and assembly and Supplementary Fig. 18. G.G. led development of protein-based SMI assays and design and interpretation of protein experiments. M.T.G. analyzed profiling data of human lung cells. M.L.H. contributed to the development of reporters and analysis of FFPE RNA quality and created Supplementary Fig. 6. R.K. designed and developed reporter structure. E.E.K. contributed to SMI methods. D.K. developed the overall concept, designed and guided experiments and analyzed data. T.K.K. supervised data collection and interpretation of human lung samples. Y.K. supervised data collection, analysis and interpretation of human lung samples. A. Klock developed the SMI ISH probe design pipeline and designed SMI ISH probes used in the 980-plex RNA panel. M.K. developed the primary data analysis pipeline for RNA and protein targets. A. Kutchma designed SMI ISH probes and wrote the section SMI ISH probe design. Z.R.L. designed and developed the protein assay, executed protein analyses and performed manuscript writing and editing. Y.L. conducted a pathological review to identify the correct staining pattern for all antibodies used. J.S.N.’s team was responsible for all chemistry process development efforts and supply chain management pertaining to the outsourcing of oligonucleotide synthesis, process improvements, price negotiation and supply agreement, key component scale-up and validation of all custom reagents and DNA components in R&D required for SMI—including contract manufacturing operation and validation of the PC spacer, synthesis and quality control of the three large-scale component sets needed for SMI reporter manufacturing plus high-throughput synthesis, outsourcing, quality control and processing of the thousands of required ISH probes. G.T.O. developed morphology and segmentation markers. E.P.P. developed the SMI instrument optical subsystem, instrument validation and support. J.C.P. developed the encoding scheme, screened reporter sequences and developed readout sequences. T.P.-E. optimized protein assay and undertook data collection, protein analyses and manuscript writing/editing. E.P. designed the 980-plex RNA panel with P.D. and contributed to manuscript writing/editing. T.R. developed the secondary analysis and target decoding pipeline for RNA targets, and the SMI instrument fluidic subsystem and workflow software. Z.R. developed the LR interaction analysis method, analyzed LR interactions across all tumors and contributed to manuscript writing/editing. M.R. performed initial chemistry development, supervised data release and contributed to writing the manuscript. A.R. was responsible for SMI protein assay content design and reagent validation. D.R. created plots of transcript positions overlaid on morphology and segmentation images for the figures. H.S. carried out manuscript development, writing and editing and created figures and tables. A.W.W. designed and developed the cell segmentation pipeline and contributed to manuscript writing/editing. C.A.W.-W. performed lung RNA isolation, processing and quality measurement and NGS library preparation and sequencing. L.W. established the cell segmentation pipeline, optimized the on-instrument SMI readout workflow and contributed to manuscript writing/editing. J.M.B. conceived the project, helped with experimental design and analysis and contributed to manuscript writing/editing.

Corresponding author

Correspondence to Joseph M. Beechem.

Ethics declarations

Competing interests

All authors are employees of NanoString Technologies and hold NanoString stock or stock options. D.K. is an employee of Dxome Co.

Peer review

Peer review information

Nature Biotechnology thanks Sanjay Tyagi and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Supplementary information

Supplementary Information

Supplementary Figs. 1–18.

Reporting Summary

Supplementary Tables 1–11.

Supplementary Tables 1–11; combined tables separated by tab.

Rights and permissions

Springer Nature or its licensor holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

He, S., Bhatt, R., Brown, C. et al. High-plex imaging of RNA and proteins at subcellular resolution in fixed tissue by spatial molecular imaging. Nat Biotechnol 40, 1794–1806 (2022). https://doi.org/10.1038/s41587-022-01483-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-022-01483-z

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing