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:

Prediction of single-cell RNA expression profiles in live cells by Raman microscopy with Raman2RNA

Abstract

Single-cell RNA sequencing and other profiling assays have helped interrogate cells at unprecedented resolution and scale, but are inherently destructive. Raman microscopy reports on the vibrational energy levels of proteins and metabolites in a label-free and nondestructive manner at subcellular spatial resolution, but it lacks genetic and molecular interpretability. Here we present Raman2RNA (R2R), a method to infer single-cell expression profiles in live cells through label-free hyperspectral Raman microscopy images and domain translation. We predict single-cell RNA sequencing profiles nondestructively from Raman images using either anchor-based integration with single molecule fluorescence in situ hybridization, or anchor-free generation with adversarial autoencoders. R2R outperformed inference from brightfield images (cosine similarities: R2R >0.85 and brightfield <0.15). In reprogramming of mouse fibroblasts into induced pluripotent stem cells, R2R inferred the expression profiles of various cell states. With live-cell tracking of mouse embryonic stem cell differentiation, R2R traced the early emergence of lineage divergence and differentiation trajectories, overcoming discontinuities in expression space. R2R lays a foundation for future exploration of live genomic dynamics.

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: R2R.
Fig. 2: R2R accurately distinguishes cell types and predicts binary expression of marker genes in a mixture of mouse fibroblasts and iPS cells.
Fig. 3: R2R predicts single-cell RNA profiles during reprogramming of mouse fibroblasts to iPS cells.
Fig. 4: R2R tracks and predicts gene expression dynamics in live single cells during mES cell differentiation.

Similar content being viewed by others

Data availability

Raw data for the mouse reprogramming scRNA-seq are available for download from NCBI Gene Expression Omnibus (https://www.ncbi.nlm.nih.gov/geo) under project number: GSE122662. mES cell differentiation scRNA-seq data are available publicly20. All other data are available at https://console.cloud.google.com/storage/browser/raman2rna.

Code availability

Code for R2R are available at https://github.com/kosekijkk/raman2rna. Control software for the multimodal Raman microscope are available at https://github.com/kosekijkk/multimodal-raman-acq.

References

  1. Tanay, A. & Regev, A. Scaling single-cell genomics from phenomenology to mechanism. Nature 541, 331–338 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Schiebinger, G. et al. Optimal-transport analysis of single-cell gene expression identifies developmental trajectories in reprogramming. Cell 176, 928–943.e22 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. La Manno, G. et al. RNA velocity of single cells. Nature 560, 494–498 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Bergen, V., Lange, M., Peidli, S., Wolf, F. A. & Theis, F. J. Generalizing RNA velocity to transient cell states through dynamical modeling. Nat. Biotechnol. 38, 1408–1414 (2020).

    Article  CAS  PubMed  Google Scholar 

  6. Wagner, D. E. & Klein, A. M. Lineage tracing meets single-cell omics: opportunities and challenges. Nat. Rev. Genet. 21, 410–427 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Gorin, G., Fang, M., Chari, T. & Pachter, L. RNA velocity unraveled. Preprint at bioRxiv https://doi.org/10.1101/2022.02.12.480214 (2022). 2022.02.12.480214 .

  8. Rabani, M. et al. High-resolution sequencing and modeling identifies distinct dynamic RNA regulatory strategies. Cell 159, 1698–1710 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Wei, L. et al. Super-multiplex vibrational imaging. Nature 544, 465–470 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Chen, W. et al. Live-seq enables temporal transcriptomic recording of single cells. Nature https://doi.org/10.1038/s41586-022-05046-9 (2022).

  11. Kobayashi-Kirschvink, K. J. et al. Linear regression links transcriptomic data and cellular Raman spectra. Cell Syst. https://doi.org/10.1016/j.cels.2018.05.015 (2018).

  12. Singh, S. P. et al. Label-free characterization of ultra violet-radiation-induced changes in skin fibroblasts with Raman spectroscopy and quantitative phase microscopy. Sci. Rep. 7, 10829 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ichimura, T. et al. Visualizing cell state transition using Raman spectroscopy. PLoS ONE 9, e84478 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  14. Ho, C.-S. et al. Rapid identification of pathogenic bacteria using Raman spectroscopy and deep learning. Nat. Commun. 10, 4927 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Biancalani, T. et al. Deep learning and alignment of spatially resolved single-cell transcriptomes with Tangram. Nat. Methods 18, 1352–1362 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Stadtfeld, M., Maherali, N., Borkent, M. & Hochedlinger, K. A reprogrammable mouse strain from gene-targeted embryonic stem cells. Nat. Methods 7, 53–55 (2010).

    Article  CAS  PubMed  Google Scholar 

  17. Choi, H. M. T. et al. Third-generation in situ hybridization chain reaction: multiplexed, quantitative, sensitive, versatile, robust. Development 145, dev165753 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  18. McInnes, L., Healy, J., Saul, N. & Großberger, L. UMAP: uniform manifold approximation and projection. J. Open Source Softw. 3, 861 (2018).

    Article  Google Scholar 

  19. Germond, A., Panina, Y., Shiga, M., Niioka, H. & Watanabe, T. M. Following embryonic stem cells, their differentiated progeny, and cell-state changes during iPS reprogramming by Raman spectroscopy. Anal. Chem. 92, 14915–14923 (2020).

    Article  CAS  PubMed  Google Scholar 

  20. Semrau, S. et al. Dynamics of lineage commitment revealed by single-cell transcriptomics of differentiating embryonic stem cells. Nat. Commun. 8, 1096 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Zhou, Y. et al. Metascape provides a biologist-oriented resource for the analysis of systems-level datasets. Nat. Commun. 10, 1523 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  22. Lange, M. et al. CellRank for directed single-cell fate mapping. Nat. Methods 19, 159–170 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Li, C., Virgilio, M., Collins, K. & Welch, J. D. Multi-omic single-cell velocity models epigenome–transcriptome interactions and improves cell fate prediction. Nat. Biotechnol. 41, 387–398 (2023).

  24. Wu, H. et al. In vivo lipidomics using single-cell Raman spectroscopy. Proc. Natl Acad. Sci. USA 108, 3809–3814 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Kunapareddy, N., Freyer, J. P. & Mourant, J. R. Raman spectroscopic characterization of necrotic cell death. J. Biomed. Opt. 13, 054002 (2008).

    Article  PubMed  Google Scholar 

  26. Freudiger, C. W. et al. Label-free biomedical imaging with high sensitivity by stimulated Raman scattering microscopy. Science 322, 1857–1861 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Bai, Y. et al. Ultrafast chemical imaging by widefield photothermal sensing of infrared absorption. Sci. Adv. 5, eaav7127 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tamamitsu, M., Toda, K., Horisaki, R. & Ideguchi, T. Quantitative phase imaging with molecular vibrational sensitivity. Opt. Lett. 44, 3729–3732 (2019).

    Article  CAS  PubMed  Google Scholar 

  29. Cleary, B., Cong, L., Cheung, A., Lander, E. S. & Regev, A. Efficient generation of transcriptomic profiles by random composite measurements. Cell 171, 1424–1436.e18 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  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  PubMed  PubMed Central  Google Scholar 

  32. Wang, X. et al. Three-dimensional intact-tissue sequencing of single-cell transcriptional states. Science https://doi.org/10.1126/science.aat5691 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Alon, S. et al. Expansion sequencing: spatially precise in situ transcriptomics in intact biological systems. Science 371, eaax2656 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Kang, J. W., Nguyen, F. T. & Lue, N. Temporal imaging of live cells by high-speed confocal Raman microscopy. Materials 14, 3732 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Kang, J. W., So, P. T. C., Dasari, R. R. & Lim, D.-K. High resolution live cell Raman imaging using subcellular organelle-targeting SERS-sensitive gold nanoparticles with highly narrow intra-nanogap. Nano Lett. 15, 1766–1772 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Radford, A. et al. Learning transferable visual models from natural language supervision. In International Conference on Machine Learning 8748–8763 (PMLR, 2021).

  37. Edelstein, A., Amodaj, N., Hoover, K., Vale, R. & Stuurman, N. Computer control of microscopes using µManager. Curr. Protoc. Mol. Biol. https://doi.org/10.1002/0471142727.mb1420s92 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902.e21 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  39. Polo, J. M. et al. A molecular roadmap of reprogramming somatic cells into iPS cells. Cell 151, 1617–1632 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. opencv. GitHub https://github.com/opencv/opencv (2023).

  41. rampy. GitHub https://github.com/charlesll/rampy (2021).

  42. biomagdsb. GitHub https://github.com/spreka/biomagdsb (2020).

  43. Prokhorenkova, L., Gusev, G., Vorobev, A., Dorogush, A.V. & Gulin, A. CatBoost: unbiased boosting with categorical features. In Advances in Neural Information Processing Systems 31 (eds Bengio, S. et al.) (NeurIPS, 2018).

  44. Yang, K. D. et al. Multi-domain translation between single-cell imaging and sequencing data using autoencoders. Nat. Commun. 12, 31 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. pegasus. GitHub https://github.com/klarman-cell-observatory/pegasus (2022).

  46. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Ounkomol, C., Seshamani, S., Maleckar, M. M., Collman, F. & Johnson, G. R. Label-free prediction of three-dimensional fluorescence images from transmitted-light microscopy. Nat. Methods 15, 917–920 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Weigert, M., Schmidt, U., Haase, R., Sugawara, K. & Myers, G. Star-convex Polyhedra for 3D object detection and segmentation in microscopy. In Proceedings of the IEEE Winter Conference on Applications of Computer Vision (WACV) 3655–3662 (IEEE, 2020).

  49. napari. GitHub https://github.com/napari/napari (2022).

  50. Ulicna, K., Vallardi, G., Charras, G. & Lowe, A. R. Automated deep lineage tree analysis using a bayesian single cell tracking approach. Front. Comput. Sci. 3, 734559 (2021).

  51. Parekh, S., Ziegenhain, C., Vieth, B., Enard, W. & Hellmann, I. zUMIs—a fast and flexible pipeline to process RNA sequencing data with UMIs. Gigascience 7, giy059 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work is part of the Human Cell Atlas. We thank L. Gaffney and A. Hupalowska for graphical editing of figures and E. Lander, R. Jaenisch, D. Hekstra and J. Kirschvink for their helpful discussion and insights. K.J.K.-K. was supported by the Japan Society for the Promotion of Science Postdoctoral Fellowship for Overseas Researchers, and the Naito Foundation Overseas Postdoctoral Fellowship. B.G. was supported by the MathWorks Fellowship. J.S. was supported by the Helen Hay Whitney Foundation and NIH, and funds from the Broad Institute of MIT and Harvard and Massachusetts General Hospital. This research was funded by NIH National Institute of Biomedical Imaging and Bioengineering, grant P41EB015871 (J.W.K. and P.T.C.S.), NIH grant UG3 UG3CA275687 (J.W.K., P.T.C.S. and J.S.), NIH grant U19 MH114821 (A.R. and T.B.), HubMap UH3CA246632 (A.R. and T.B.), 4R00HD096049-03 (J.S.), 5R00HD096049-04 (J.S.) and HHMI and the Klarman Cell Observatory (A.R.). A.R. was a Howard Hughes Medical Institute Investigator when this work was initiated.

Author information

Authors and Affiliations

Authors

Contributions

K.J.K.-K. and A.R. conceived the research. K.J.K.-K., J.S., T.B. and A.R. developed the methodology. J.S., T.B., P.T.C.S. and A.R. funded and supervised research. K.J.K.-K., J.S. and J.R.O. performed reprogramming and differentiation experiments. K.J.K.-K. developed the Raman microscope and control software with supervision from J.W.K. and P.T.C.S. K.J.K.-K., E.I.G. and K.Z. performed smFISH. K.J.K.-K., S.G., T.J. and T.B. developed the Raman spectral preprocessing and classification pipeline. K.J.K.-K. developed the image registration, cell tracking, anchor-based inference and feature importance analysis pipeline. K.J.K.-K., C.S.C. and B.G. performed brightfield-based classification and regression. C.S.C. and K.J.K.-K. designed fully connected neural nets and AAEs with guidance from A.R. K.J.K.-K., J.S. and A.R. wrote the manuscript with input from all the authors.

Corresponding authors

Correspondence to Koseki J. Kobayashi-Kirschvink, Tommaso Biancalani, Jian Shu or Aviv Regev.

Ethics declarations

Competing interests

A.R. is a co-founder and equity holder of Celsius Therapeutics, an equity holder in Immunitas, and was a scientific advisory board member of ThermoFisher Scientific, Syros Pharmaceuticals, Neogene Therapeutics and Asimov until 31 July 2020. A.R. is an employee of Genentech from 1 August 2020 with equity in Roche. T.B., S.G. and T.J. are employees of Genentech from 1 Feburary 2021, 29 March 2021 and 5 June 2023, respectively. J.S. is a scientific advisor for Arcadia Science. A patent application has been filed by the Broad Institute related to this work. The other authors declare no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Carsten Marr 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.

Extended data

Extended Data Fig. 1 A multi-modal Raman microscope capable of fluorescence imaging and Raman microscopy.

Schematic of a Raman microscope integrated with a wide-field fluorescence microscope for simultaneous detection of nuclei staining, bright field, fluorescence channels, and Raman images.

Extended Data Fig. 2 Raman-predicted and scRNA-seq measured pseudo-bulk profiles are well correlated across cell types.

scRNA-seq measured (y axis) and R2R-predicted (x axis) expression for each gene (dot) in pseudo-bulk RNA profiles averaged across cells labeled as iPSC (top left), epithelial (top right), stromal (bottom left) and MET (bottom right). Cosine similarity is denoted at the top left corner.

Extended Data Fig. 3 Measured and Raman-predicted single cell profiles co-embed well as reflected by gene scores for each cell type.

UMAP co-embedding of Raman predicted RNA profiles and measured scRNA-seq profiles (dots) colored by scores of marker gene for different cell types (rows) determined by smFISH measurements (left, for cells with Raman-predicted profiles) or real scRNA-seq measurements (right, for cells with scRNA-seq profiles).

Extended Data Fig. 4 Measured and Raman-predicted single cell profiles co-embed well as reflected by smFISH measurement of Raman cells.

UMAP co-embedding of Raman predicted RNA profiles and measured scRNA-seq profiles (dots) where the Raman cells are colored by smFISH measurement of each of nine anchor genes.

Extended Data Fig. 5 Measured and Raman-predicted single cell profiles co-embed well as reflected by scRNA-seq based expression of nine anchor genes.

UMAP co-embedding of Raman predicted RNA profiles and measured scRNA-Seq profiles (dots) where the scRNA-seq profiled cells are colored by scRNA-seq measured expression of each of nine anchor genes.

Extended Data Fig. 6 Distributions of expression of marker genes based on R2R-predicted profiles.

Distributions (density plots) of the predicted expression in Raman2RNA inferred profiles for each marker gene (panel) in its expected corresponding cell type (blue, based on the predicted expression profiles) and all other cells (orange).

Extended Data Fig. 7 Distributions of expression of marker genes based on real smFISH profiles.

Distributions (density plots) of the real smFISH profiles for each marker gene (panel) in its expected corresponding cell type (blue, based on the R2R predicted expression profiles) and all other cells (orange).

Extended Data Fig. 8 Raman spectral feature importance scores for each smFISH anchor gene and its average across all genes for a cell type.

Feature importance scores (y axis) for marker genes of each cell type (top two rows), and for all cell types (bottom row), along the Raman spectrum (x axis). Known signals2 are annotated in the top left panel (identical to Fig. 3j).

Extended Data Fig. 9 Adversarial autoencoder (AAE) based model for anchor-free generation of scRNA-seq from Raman profiles.

Top: Two autoencoders (AEs) – one for Raman and the other for scRNA-seq – are trained adversarially to learn two indistinguishable latent spaces. Once a common latent space is found, new Raman spectra are encoded using the encoder part of the Raman AE and decoded to scRNA-seq using the decoder part of the scRNA-seq AE (bottom).

Extended Data Fig. 10 Anchor-free R2R profiles capture variance in single cell profiles as indicated by co-embedding.

UMAP co-embedding of anchor-free R2R-generated profiles and real scRNA-seq (dots) colored by cell types determined by Tangram label-transfer on smFISH measurements (left, for cells with R2R-generated profiles) or by ground truth scRNA-seq (right, for cells with scRNA-seq profiles).

Supplementary information

Supplementary Information

Supplementary Figs. 1–19 and Note.

Reporting Summary

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) 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

Kobayashi-Kirschvink, K.J., Comiter, C.S., Gaddam, S. et al. Prediction of single-cell RNA expression profiles in live cells by Raman microscopy with Raman2RNA. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-023-02082-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-023-02082-2

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