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.

  • Analysis
  • Published:

Bayesian inference of gene expression states from single-cell RNA-seq data

Abstract

Despite substantial progress in single-cell RNA-seq (scRNA-seq) data analysis methods, there is still little agreement on how to best normalize such data. Starting from the basic requirements that inferred expression states should correct for both biological and measurement sampling noise and that changes in expression should be measured in terms of fold changes, we here derive a Bayesian normalization procedure called Sanity (SAmpling-Noise-corrected Inference of Transcription activitY) from first principles. Sanity estimates expression values and associated error bars directly from raw unique molecular identifier (UMI) counts without any tunable parameters. Using simulated and real scRNA-seq datasets, we show that Sanity outperforms other normalization methods on downstream tasks, such as finding nearest-neighbor cells and clustering cells into subtypes. Moreover, we show that by systematically overestimating the expression variability of genes with low expression and by introducing spurious correlations through mapping the data to a lower-dimensional representation, other methods yield severely distorted pictures of the data.

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: Summary of the Sanity approach.
Fig. 2: Effects of Poisson fluctuations on gene expression variance.
Fig. 3: Accuracy of gene expression estimates as a function of depth of coverage.
Fig. 4: Correlations between inferred gene expression levels and library size and between pairs of genes.
Fig. 5: Accuracy of the k nearest-neighbor and clustering predictions.

Similar content being viewed by others

Data availability

The raw UMI count tables for each of the scRNA-seq datasets as well as all normalized expression values as inferred by each of the methods are freely available from https://doi.org/10.5281/zenodo.4009187.

Code availability

Sanity was implemented in C and is freely available for download at https://github.com/jmbreda/Sanity. Besides Sanity itself, we also provide code for estimating pairwise distances between cells. In addition, at the same GitHub site, we provide a collection of scripts and supplementary files that should allow other researchers to reproduce the results presented in this publication.

References

  1. Picelli, S. et al. Smart-seq2 for sensitive full-length transcriptome profiling in single cells. Nat. Methods 10, 1096–1098 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-seq: single-cell RNA-seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Buenrostro, J. D. et al. Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523, 486–490 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Cusanovich, D. A. et al. Multiplex single cell profiling of chromatin accessibility by combinatorial cellular indexing. Science 348, 910–914 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rotem, A. et al. Single-cell ChIP–seq reveals cell subpopulations defined by chromatin state. Nat. Biotechnol. 33, 1165–1172 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Smallwood, S. A. et al. Single-cell genome-wide bisulfite sequencing for assessing epigenetic heterogeneity. Nat. Methods 11, 817–820 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Nagano, T. et al. Single-cell Hi-C reveals cell-to-cell variability in chromosome structure. Nature 502, 59–64 (2013).

    Article  CAS  PubMed  Google Scholar 

  10. McKenna, A. et al. Whole-organism lineage tracing by combinatorial and cumulative genome editing. Science 353, aaf7907 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  11. Kalhor, R. et al. Developmental barcoding of whole mouse via homing CRISPR. Science 361, eaat9804 (2018).

  12. Frieda, K. L. et al. Synthetic recording and in situ readout of lineage information in single cells. Nature 541, 107–111 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Frei, A. P. et al. Highly multiplexed simultaneous detection of RNAs and proteins in single cells. Nat. Methods 13, 269–275 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Raj, B. et al. Simultaneous single-cell profiling of lineages and cell types in the vertebrate brain. Nat. Biotechnol. 36, 442–450 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Spanjaard, B. et al. Simultaneous lineage tracing and cell-type identification using CRISPR–Cas9-induced genetic scars. Nat. Biotechnol. 36, 469–473 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Wagner, D. E. et al. Single-cell mapping of gene expression landscapes and lineage in the zebrafish embryo. Science 360, 981–987 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Angermueller, C. et al. Parallel single-cell sequencing links transcriptional and epigenetic heterogeneity. Nat. Methods 13, 229–232 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Clark, S. J. et al. scNMT-seq enables joint profiling of chromatin accessibility DNA methylation and transcription in single cells. Nat. Commun. 9, 781 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  19. Adamson, B. et al. A multiplexed single-cell CRISPR screening platform enables systematic dissection of the unfolded protein response. Cell 167, 1867–1882 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Dixit, A. et al. Perturb-seq: dissecting molecular circuits with scalable single-cell RNA profiling of pooled genetic screens. Cell 167, 1853–1866 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Jaitin, D. A. et al. Dissecting immune circuits by linking CRISPR-pooled screens with single-cell RNA-seq. Cell 167, 1883–1896 (2016).

    Article  CAS  PubMed  Google Scholar 

  22. Datlinger, P. et al. Pooled CRISPR screening with single-cell transcriptome readout. Nat. Methods 14, 297–301 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Regev, A. et al. The human cell atlas. eLife 6, e27041 (2017).

  24. Rajewsky, N. et al. LifeTime and improving European healthcare through cell-based interceptive medicine. Nature 587, 377–386 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Raj, A., van den Bogaard, P., Rifkin, S. A., van Oudenaarden, A. & Tyagi, S. Imaging individual mRNA molecules using multiple singly labeled probes. Nat. Methods 5, 877–879 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Van Der Maaten, L. & Hinton, G. Visualizing data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  27. McInnes, L. & Healy, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at arXiv http://arxiv.org/abs/1802.03426 (2018).

  28. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lun, A. T., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  CAS  Google Scholar 

  30. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    Google Scholar 

  31. Huang, M. et al. SAVER: gene expression recovery for single-cell RNA sequencing. Nat. Methods 15, 539–542 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Li, W. V. & Li, J. J. An accurate and robust imputation method scImpute for single-cell RNA-seq data. Nat. Commun. 9, 997 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Grün, D., Kester, L. & Van Oudenaarden, A. Validation of noise models for single-cell transcriptomics. Nat. Methods 11, 637–640 (2014).

    Article  PubMed  CAS  Google Scholar 

  36. Brennecke, P. et al. Accounting for technical noise in single-cell RNA-seq experiments. Nat. Methods 10, 1093–1095 (2013).

    Article  CAS  PubMed  Google Scholar 

  37. Zeisel, A. et al. Cell types in the mouse cortex and hippocampus revealed by single-cell RNA-seq. Science 347, 1138–1142 (2015).

    Article  CAS  PubMed  Google Scholar 

  38. Lloyd, S. P. Least squares quantization in PCM. IEEE Trans. Inf. Theory 28, 129–137 (1982).

    Article  Google Scholar 

  39. Ward, J. H. Hierarchical grouping to optimize an objective function. J. Am. Stat. Assoc. 58, 236–244 (1963).

    Article  Google Scholar 

  40. Blondel, V. D., Guillaume, J.-L., Lambiotte, R. & Lefebvre, E. Fast unfolding of communities in large networks. J. Stat. Mech. Theory Exp. 2008, P10008 (2008).

    Article  Google Scholar 

  41. Thattai, M. Universal Poisson statistics of mRNAs with complex decay pathways. Biophys. J. 110, 301–305 (2016).

    Article  CAS  PubMed  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. 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 

  44. Padovan-Merhar, O. et al. Single mammalian cells compensate for differences in cellular volume and DNA copy number through independent global transcriptional mechanisms. Mol. Cell 58, 339–352 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Stegle, O., Teichmann, S. A. & Marioni, J. C. Computational and analytical challenges in single-cell transcriptomics. Nat. Rev. Genet. 16, 133–145 (2015).

    Article  CAS  PubMed  Google Scholar 

  46. Hoyle, D. C., Rattray, M., Jupp, R. & Brass, A. Making sense of microarray data distributions. Bioinformatics 18, 576–584 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Beal, J. Biochemical complexity drives log-normal variation in genetic expression. Eng. Biol. 1, 55–60 (2017).

    Article  Google Scholar 

  48. Love, M. I., Anders, S., Kim, V. & Huber, W. RNA-seq workflow: gene-level exploratory analysis and differential expression. F1000Res 4, 1070 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Bray, N. L., Pimentel, H., Melsted, P. & Pachter, L. Near-optimal probabilistic RNA-seq quantification. Nat. Biotechnol. 34, 525–527 (2016).

    Article  CAS  PubMed  Google Scholar 

  50. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  51. Cell Ranger DNA. https://support.10xgenomics.com/single-cell-dna/software/pipelines/latest/what-is-cell-ranger-dna

  52. Islam, S. et al. Quantitative single-cell RNA-seq with unique molecular identifiers. Nat. Methods 11, 163–166 (2014).

    Article  CAS  PubMed  Google Scholar 

  53. AlJanahi, A. A., Danielsen, M. & Dunbar, C. E. An introduction to the analysis of single-cell RNA-sequencing data. Mol. Ther. Methods Clin. Dev. 10, 189–196 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. 10X Genomics. What fraction of mRNA transcripts are captured per cell? https://kb.10xgenomics.com/hc/en-us/articles/360001539051-what-fraction-of-mrna-transcripts-are-captured-per-cell- (2018).

  55. Jaynes, E. T. Probability Theory: The Logic of Science (Cambridge Univ. Press, 2003).

  56. Svensson, V. Droplet scRNA-seq is not zero-inflated. Nat. Biotechnol. 38, 147–150 (2020).

    Article  CAS  PubMed  Google Scholar 

  57. Baron, M. et al. A single-cell transcriptomic map of the human and mouse pancreas reveals inter- and intra-cell population structure. Cell Syst. 3, 346–360 (2016).

    Google Scholar 

  58. Chen, R., Wu, X., Jiang, L. & Zhang, Y. Single-cell RNA-seq reveals hypothalamic cell diversity. Cell Rep. 18, 3227–3241 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. La Manno, G. et al. Molecular diversity of midbrain development in mouse, human, and stem cells. Cell 167, 566–580 (2016).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the Swiss National Science Foundation, grant 310030_184937. Calculations were performed at sciCORE (http://scicore.unibas.ch/), the scientific computing core facility of the University of Basel.

Author information

Authors and Affiliations

Authors

Contributions

E.v.N. developed the theoretical formalism. J.B. and E.v.N. developed the implementation and designed the benchmarking. J.B. performed all computations, analyses and simulations. J.B., M.Z. and E.v.N. interpreted the results and wrote the manuscript.

Corresponding author

Correspondence to Erik van Nimwegen.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Methods, Figs. 1–26 and Text 1.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Breda, J., Zavolan, M. & van Nimwegen, E. Bayesian inference of gene expression states from single-cell RNA-seq data. Nat Biotechnol 39, 1008–1016 (2021). https://doi.org/10.1038/s41587-021-00875-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-00875-x

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