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.

  • Brief Communication
  • Published:

Super-resolved spatial transcriptomics by deep data fusion

Abstract

Current methods for spatial transcriptomics are limited by low spatial resolution. Here we introduce a method that integrates spatial gene expression data with histological image data from the same tissue section to infer higher-resolution expression maps. Using a deep generative model, our method characterizes the transcriptome of micrometer-scale anatomical features and can predict spatial gene expression from histology images alone.

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: Overview and performance evaluation.
Fig. 2: Characterization of the transcriptome in micrometer-scale anatomical features.

Similar content being viewed by others

Data availability

The mouse olfactory bulb dataset was obtained from the Spatial Research group’s website: https://www.spatialresearch.org/resources-published-datasets/doi-10-1126science-aaf2403. The breast cancer spatial transcriptomics dataset was obtained from the 10x Genomics data repository: https://support.10xgenomics.com/spatial-gene-expression/datasets/. The breast cancer single-cell dataset was obtained from the authors of the original publication24. The squamous cell carcinoma dataset is available on Mendeley Data: https://doi.org/10.17632/2bh5fchcv6.1. The small intestine dataset is available on Mendeley Data: https://doi.org/10.17632/v8s9nz948s.1 (folder V19T26-028_B1).

Code availability

We have implemented the proposed method in PyTorch39 and the Pyro probabilistic programming language40. The code is available under the MIT license at https://github.com/ludvb/xfuse.

References

  1. Ke, R. et al. In situ sequencing for RNA analysis in preserved tissue and cells. Nat. Methods 10, 857–860 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Lee, J. H. et al. Highly multiplexed subcellular RNA sequencing in situ. Science 343, 1360–1363 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Femino, A. M. Visualization of single RNA transcripts in situ. Science 280, 585–590 (1998).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  5. 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 

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

    Article  PubMed  Google Scholar 

  7. Rodriques, S. G. et al. Slide-seq: a scalable technology for measuring genome-wide expression at high spatial resolution. Science 363, 1463–1467 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Vickovic, S. et al. High-definition spatial transcriptomics for in situ tissue profiling. Nat. Methods 16, 987–990 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Kingma, D. P. & Welling, M. Auto-encoding variational Bayes. In 2nd International Conference on Learning Representations (eds Bengio, Y. & LeCun, Y.) http://arxiv.org/abs/1312.6114 (2014).

  10. Rezende, D. J., Mohamed, S. & Wierstra, D. Stochastic backpropagation and approximate inference in deep generative models. In Proceedings of the 31th International Conference on Machine Learning http://proceedings.mlr.press/v32/rezende14.html (2014).

  11. Lein, E. S. et al. Genome-wide atlas of gene expression in the adult mouse brain. Nature 445, 168–176 (2006).

    Article  PubMed  Google Scholar 

  12. Uhlen, M. et al. Tissue-based map of the human proteome. Science 347, 1260419 (2015).

    Article  PubMed  Google Scholar 

  13. Tepe, B. et al. Single-cell RNA-seq of mouse olfactory bulb reveals cellular heterogeneity and activity-dependent molecular census of adult-born neurons. Cell Rep. 25, 2689–2703 (2018).

    Article  Google Scholar 

  14. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Bulla, R. et al. C1q acts in the tumour microenvironment as a cancer-promoting factor independently of complement activation. Nat. Commun. 7, 10346 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Metodieva, G. et al. CD74-dependent deregulation of the tumor suppressor scribble in human epithelial and breast cancer cells. Neoplasia 15, 660–668 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  18. Blei, D. M., Kucukelbir, A. & McAuliffe, J. D. Variational inference: a review for statisticians. J. Am. Stat. Assoc.112, 859–877 (2017).

  19. Lopez, R., Regier, J., Cole, M. B., Jordan, M. I. & Yosef, N. Deep generative modeling for single-cell transcriptomics. Nat. Methods 15, 1053–1058 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Eraslan, G., Simon, L. M., Mircea, M., Mueller, N. S. & Theis, F. J. Single-cell RNA-seq denoising using a deep count autoencoder. Nat. Commun. 10, 390 (2019).

  21. Kingma, D. P. & Ba, J. Adam: a method for stochastic optimization. In 3rd International Conference on Learning Representations (eds Bengio, Y. & LeCun, Y.) http://arxiv.org/abs/1412.6980 (2015).

  22. Ronneberger, O., Fischer, P. & Brox, T. U-Net: Convolutional Networks for Biomedical Image Segmentation 234–241. (Springer, 2015).

  23. Gardner, J. R., Pleiss, G., Bindel, D., Weinberger, K. Q. & Wilson, A. G. GPyTorch: blackbox matrix–matrix Gaussian process inference with GPU acceleration. In Proceedings of the 32nd International Conference on Neural Information Processing Systems (Curran Associates, 2018).

  24. Wu, S. Z. et al. A single-cell and spatially resolved atlas of human breast cancers. Nat. Genetics 53, 1334–1347 (2021).

    Article  CAS  PubMed  Google Scholar 

  25. Ji, A. L. et al. Multimodal analysis of composition and spatial architecture in human squamous cell carcinoma. Cell 182, 497–514 (2020).

    Article  Google Scholar 

  26. Macenko, M. et al. A method for normalizing histology slides for quantitative analysis. 2009 IEEE International Symposium on Biomedical Imaging: From Nano to Macro https://ieeexplore.ieee.org/document/51932500.1109/ISBI.2009.5193250 (2009).

  27. Svensson, V., Teichmann, S. A. & Stegle, O. SpatialDE: identification of spatially variable genes. Nat. Methods 15, 343–346 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Sun, S., Zhu, J. & Zhou, X. Statistical analysis of spatial expression patterns for spatially resolved transcriptomic studies. Nat. Methods 17, 193–200 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Nitzan, M., Karaiskos, N., Friedman, N. & Rajewsky, N. Gene expression cartography. Nature 576, 132–137 (2019).

    Article  CAS  PubMed  Google Scholar 

  30. Achim, K. et al. High-throughput spatial mapping of single-cell RNA-seq data to tissue of origin. Nat. Biotechnol. 33, 503–509 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Qian, X. et al. Probabilistic cell typing enables fine mapping of closely related cell types in situ. Nat. Methods 17, 101–106 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Lopez, R. et al. A joint model of unpaired data from scRNA-seq and spatial transcriptomics for imputing missing gene expression measurements. In ICML Workshop on Computational Biology (2019).

  34. Cable, D. M. et al. Robust decomposition of cell type mixtures in spatial transcriptomics. Nat. Biotechnol. https://doi.org/10.1038/s41587-021-00830-w (2021).

  35. Andersson, A. et al. Single-cell and spatial transcriptomics enables probabilistic inference of cell type topography. Commun. Biol. 3, 565 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  36. 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. 49, e50 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Stickels, R. R. et al. Highly sensitive spatial transcriptomics at near-cellular resolution with Slide-seqV2. Nat. Biotechnol. 39, 313–319 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

  39. Paszke, A. et al. PyTorch: an imperative style, high-performance deep learning library. In Advances in Neural Information Processing Systems 32 (eds Wallach, H. et al.) 8024–8035 http://papers.neurips.cc/paper/9015-pytorch-an-imperative-style-high-performance-deep-learning-library.pdf (Curran Associates, 2019).

  40. Bingham, E. et al. Pyro: deep universal probabilistic programming. J. Mach. Learn. Res. 20, 1–6 (2019).

    Google Scholar 

Download references

Acknowledgements

This work was made possible by generous support from the Knut and Alice Wallenberg Foundation, the Erling-Persson Family Foundation, the Swedish Cancer Society, the Swedish Foundation for Strategic Research, the Swedish Research Council and the Helmsley Charitable Trust.

Author information

Authors and Affiliations

Authors

Contributions

L.B. and J.M. designed the method and wrote the paper. B.H., J.B., A.A. and A.L.J. provided valuable feedback and contributed to the analyses. R.M., X.A., K.T., L.L. and N.S. performed the experiments. J.M., J.L., J.Z., P.K. and G.B. supervised the project.

Corresponding author

Correspondence to Joakim Lundeberg.

Ethics declarations

Competing interests

J.L., R.M., K.T., A.A. and L.L. are scientific consultants for 10x Genomics, which produces spatially barcoded microarrays for in situ RNA capturing. The remaining authors declare no competing interests.

Additional information

Peer review informationNature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Deconvolution experiments.

ac, Synthetic data. Receiver operating characteristic (ROC) curves for pixel-level classification of the three transcriptional subtypes: blue circle (a), red square (b), and yellow triangle (c). Dashed lines show baselines constructed by predicting the observed pixel-average in each measurement location. Ribbons indicate minima and maxima over predictions in 10 random synthetic image patches. df, Biological data. d, The ground truth expression data is downsampled by merging neighboring measurement locations and summing their count values XA + XB = X. The model is trained on the downsampled data X and used to predict the component counts XA and XB for each gene. e, Predicted direction against ground truth for observations with a 95% credibility of one component having a strictly higher expression than the other. Points show the medians of the predictive distributions, and error bars indicate 90% credibility intervals. Colors indicate if the predicted direction is the same as the ground truth direction. The dashed line indicates identity. For readability, points are only shown for the 10 highest-expressed genes. Hexagonal bins show observations for all genes (n = 12 776). Statistics are based on all genes. f, Directional misprediction against prediction uncertainty. In red, points indicate the 50th and error bars the 5th and 95th percentiles in evenly distributed bins.

Extended Data Fig. 2 Comparison of inferred super-resolved expression maps to in situ hybridization reference data.

Random samples from the 1000 highest-expressed genes. Raw: Raw expression data (Voronoi tesselation). Inferred: Inferred super-resolved expression maps. ISH: In situ hybridization reference data from the Allen Mouse Brain Atlas11. Images show a representative sample of the 12 mouse olfactory bulb sections in the dataset.

Extended Data Fig. 3 Prediction of spatial gene expression from histology images, mouse olfactory bulb experiments.

a, Histology image of holdout section (hematoxylin and eosin stain). b, Summarized expression map of the predicted metagene expression in the holdout section. c, Comparison of summarized expression maps constructed from normalized log ground truth gene expression in the held-out section (top) and normalized log predicted gene expression at the ground truth measurement locations using data from b (bottom). Results are based on an analysis that uses the 12th sample as holdout section (shown in a–c) and the remaining 11 samples as reference experiments.

Extended Data Fig. 4 Prediction of spatial gene expression from histology images, squamous cell carcinoma experiments.

The dataset consists of four serial tissue sections spaced 150 μm apart. The outer sections A and D are used as reference experiments to predict expression in the intermediate holdout sections B and C. a, Top: Histological image data (hematoxylin and eosin stains). Middle: Predicted summarized expression maps. Bottom: Predicted expression against ground truth for all genes (n = 11 025) in 100 randomly sampled test locations. b,c, Baseline experiments. b, Constant prediction against ground truth for all genes in the same test locations as in (a). For each gene, the prediction is the mean expression in sections A and D. c, Image intensity-based linear regression prediction against ground truth for all genes in the same test locations as in (a). The expression Xlg of gene g in location l is modeled as \({X}_{lg}={\beta }_{g}^{0}+{\beta }_{g}{I}_{l}+{\epsilon }_{lg}\), where Il is a vector of the channel-wise 5-binned image intensities of location l and ϵlg a standard normal noise term. The parameters \({\beta }_{g}^{0}\) and βg are selected by maximum likelihood estimation with data from sections A and D. Predictions are given by \({X}_{lg}^{* }=\max (0,\beta^0_g+{\beta }_{g}{I}_{l})\). d, Stability to variation in staining intensities. Left: Evaluated hematoxylin (H) and eosin (E) concentrations. Mixes are produced synthetically by rescaling the H and E channels (Methods). Images show representative close-ups from one of the four sections in the dataset. Right: Gene-wise Pearson correlation over all test locations in each holdout section evaluated on the n = 100 highest-expressed genes. Boxes show 25th, 50th, and 75th percentiles. Outliers are represented by points and defined as observations further than 1.5 interquartile ranges from the hinges. Whiskers indicate the extent of all non-outlier observations.

Extended Data Fig. 5 Run-to-run variability.

Results are based on four restarts of an analysis of the ileum of the human small intestine. a, Correlation plots. Each plot shows the predicted mean count for every gene in 100 test regions in two different runs. The test regions are sampled uniformly over the tissue surface and have the same size as the measurement locations in the original dataset. Correlation coefficients are computed over all genes and test locations (n = 6869 × 100 = 686 900). b, Differences in predicted means E[νi] − E[νj] against prediction uncertainty \(\sqrt{V({\nu }_{i})+V({\nu }_{j})}\) for runs i and j. In red, points indicate the 50th and error bars the 5th and 95th percentiles in evenly distributed bins.

Extended Data Fig. 6 Robustness to measurement location misalignment.

a, Hematoxylin and eosin stain of a section from the ileum of the human small intestine. Representative close-up of a small area of the brush border. The brush border in the section measures approximately 7 mm in length. b, Conceptual illustration of the measurement locations on the Visium array. Dark circles indicate test locations withheld during training. Light circles indicate training locations over three misalignment levels: 0.0 (light green), 1.0 (green), and 2.0 (blue) radii of the measurement locations (r = 55 μm). The direction of the misalignment is uniformly random. c,d, Gene-wise Pearson correlation between predicted and ground truth expression (c) and coefficient of determination (d) over the test locations of the n = 100 highest-expressed genes for increasing offsets. Boxes show 25th, 50th, and 75th percentiles. Outliers are represented by points and defined as observations further than 1.5 interquartile ranges from the hinges. Whiskers indicate the extent of all non-outlier observations. Pairwise p-values are based two-sided Wilcoxon signed-rank tests. Exact p-values (top to bottom): c, 2.98 × 10−7 and 8.30 × 10−1; d, 3.45 × 10−9 and 8.14 × 10−1. eg, Predicted expression of CDHR5 when the training set has 0.0 (e), 1.0 (f), or 2.0 (g) radii misalignment. Close-ups of the same area as in a. h, Reference antibody staining for CDHR5 in the small intestine from the Human Protein Atlas12.

Extended Data Fig. 7 Robustness to image disruptions.

a, Hematoxylin and eosin stains of a section from the ileum of the human small intestine with increasing levels of occlusion noise (ε). Representative close-ups of the smooth muscle layers. The smooth muscle layers in the section measure approximately 3 × 5 mm2. Noise is added by randomly sampling a proportion ε of tiles from a 100 × 100 grid covering the histology image and replacing them with the mean color intensity of the slide. b, Conceptual illustration of the measurement locations on the Visium array. The locations are divided into a training (light green) and test set. The test set is further divided into regions that are fully visible in all experiments (green) and regions that are at least partially occluded in some experiments (blue). ch, Performance under different occlusion levels evaluated using the gene-wise Pearson correlation between predicted and ground truth expression (c–e) and coefficient of determination (f–h) over the test locations of the n = 100 highest-expressed genes. Boxes show 25th, 50th, and 75th percentiles. Outliers are represented by points and defined as observations further than 1.5 interquartile ranges from the hinges. Whiskers indicate the extent of all nonoutlier observations. Pairwise p-values are based two-sided Wilcoxon signed-rank tests. Exact p-values (top to bottom): c, 1.48 × 10−17 and 9.18 × 10−18; d, 1.71 × 10−13 and 3.77 × 10−13; e, 9.75 × 10−18 and 8.39 × 10−18; f, 8.65 × 10−18 and 5.85 × 10−18; g, 1.15 × 10−16 and 2.24 × 10−15; h, 7.44 × 10−18 and 6.03 × 10−18. i, Prediction of ACTG2, a gene coding for gamma-enteric smooth muscle actin, over different occlusion levels.

Extended Data Fig. 8 Differential gene expression and cell-type composition.

a,b, Differential gene expression, mouse olfactory bulb dataset. a, Annotation of the mitral cell layer (MCL). Percentages indicate area overlap with pixel annotation. b, Agreement with MCL marker reference list13 over different set sizes of genes predicted to be differentially expressed. Genes are ranked by the inverted coefficient of variation of their posterior log fold change (Super-resolved ST) or p-value (Seurat). Only genes with a predicted positive log fold change are shown. ce, Cell-type composition, ductal carcinoma in situ (DCIS) dataset. c, Predicted cell types in each measurement location. Colors correspond to the score-weighted sums of the cell-type labels’ RGB coordinates. d, Measurement-level classification as a function of proximity to the tumor edge. Bar heights show classification scores across all measurement locations weighted by their overlap with each distance isoline. e, Pixel-level classification as a function of proximity to the tumor edge. Bar heights show classification scores based on the predicted expression for the pixel band at each distance. Red dashed line: Tumor edge. Black dotted line: 200 μm isoline.

Extended Data Fig. 9 Architecture.

a, Fusion network. b, Image data decoder. c, Expression data decoder. Volume dimensions and number of down- and upsampling steps are exemplative.

Extended Data Fig. 10 Runtime.

Normalized ELBO over time for three runs with varying dataset sizes. Vertical lines indicate time points when the runs reached 95% of the maximum attained normalized ELBO for the first time.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Bergenstråhle, L., He, B., Bergenstråhle, J. et al. Super-resolved spatial transcriptomics by deep data fusion. Nat Biotechnol 40, 476–479 (2022). https://doi.org/10.1038/s41587-021-01075-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-01075-3

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