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:

Single-cell imaging and RNA sequencing reveal patterns of gene expression heterogeneity during fission yeast growth and adaptation

Abstract

Phenotypic cell-to-cell variability is a fundamental determinant of microbial fitness that contributes to stress adaptation and drug resistance. Gene expression heterogeneity underpins this variability but is challenging to study genome-wide. Here we examine the transcriptomes of >2,000 single fission yeast cells exposed to various environmental conditions by combining imaging, single-cell RNA sequencing and Bayesian true count recovery. We identify sets of highly variable genes during rapid proliferation in constant culture conditions. By integrating single-cell RNA sequencing and cell-size data, we provide insights into genes that are regulated during cell growth and division, including genes whose expression does not scale with cell size. We further analyse the heterogeneity of gene expression during adaptive and acute responses to changing environments. Entry into the stationary phase is preceded by a gradual, synchronized adaptation in gene regulation that is followed by highly variable gene expression when growth decreases. Conversely, sudden and acute heat shock leads to a stronger, coordinated response and adaptation across cells. This analysis reveals that the magnitude of global gene expression heterogeneity is regulated in response to different physiological conditions within populations of a unicellular eukaryote.

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: Imaging and transcriptome analysis of single fission yeast cells.
Fig. 2: Cell-to-cell variability of the fission yeast transcriptome.
Fig. 3: Cell-size dependence of the fission yeast transcriptome.
Fig. 4: Gene-expression heterogeneity of fission yeast populations in response to environmental changes.

Similar content being viewed by others

Data availability

All raw scRNA-seq datasets are available in ArrayExpress accession number E-MTAB-6825. Cell size measurement and all smFISH data are available as Supplementary Material. The bayNorm package is available from Bioconductor: https://bioconductor.org/packages/release/bioc/html/bayNorm.html. All figures except Fig. 1a and Supplementary Fig. 2a contain original data.

References

  1. Kaern, M., Elston, T. C., Blake, W. J. & Collins, J. J. Stochasticity in gene expression: from theories to phenotypes. Nat. Rev. Genet. 6, 451–464 (2005).

    Article  CAS  PubMed  Google Scholar 

  2. Shahrezaei, V. & Swain, P. S. The stochastic nature of biochemical networks. Curr. Opin. Biotechnol. 19, 369–374 (2008).

    Article  CAS  PubMed  Google Scholar 

  3. Raj, A. & van Oudenaarden, A. Nature, nurture, or chance: stochastic gene expression and its consequences. Cell 135, 216–226 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Elowitz, M. B., Levine, A. J., Siggia, E. D. & Swain, P. S. Stochastic gene expression in a single cell. Science 297, 1183–1186 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Segal, E. & Widom, J. From DNA sequence to transcriptional behaviour: a quantitative approach. Nat. Rev. Genet. 10, 443–456 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Battich, N., Stoeger, T. & Pelkmans, L. Control of transcript variability in single mammalian cells. Cell 163, 1596–1610 (2015).

    Article  CAS  PubMed  Google Scholar 

  7. Shahrezaei, V. & Marguerat, S. Connecting growth with gene expression: of noise and numbers. Curr. Opin. Microbiol. 25, 127–135 (2015).

    Article  CAS  PubMed  Google Scholar 

  8. Mellor, J. The molecular basis of metabolic cycles and their relationship to circadian rhythms. Nat. Struct. Mol. Biol. 23, 1035–1044 (2016).

    Article  CAS  PubMed  Google Scholar 

  9. Svensson, V., Vento-Tormo, R. & Teichmann, S. A. Exponential scaling of single-cell RNA-seq in the past decade. Nat. Protoc. 13, 599–604 (2018).

    Article  CAS  PubMed  Google Scholar 

  10. Stubbington, M. J. T., Rozenblatt-Rosen, O., Regev, A. & Teichmann, S. A. Single-cell transcriptomics to explore the immune system in health and disease. Science 358, 58–63 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Baslan, T. & Hicks, J. Unravelling biology and shifting paradigms in cancer with single-cell sequencing. Nat. Rev. Cancer 17, 557–569 (2017).

    Article  CAS  PubMed  Google Scholar 

  12. Griffiths, J. A., Scialdone, A. & Marioni, J. C. Using single-cell genomics to understand developmental processes and cell fate decisions. Mol. Syst. Biol. 14, e8046 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Saliba, A.-E., C Santos, S. & Vogel, J. New RNA-seq approaches for the study of bacterial pathogens. Curr. Opin. Microbiol. 35, 78–87 (2017).

    Article  CAS  PubMed  Google Scholar 

  14. Gasch, A. P. et al. Single-cell RNA sequencing reveals intrinsic and extrinsic regulatory heterogeneity in yeast responding to stress. PLoS Biol. 15, e2004050 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  15. Soumillon, M., Cacchiarelli, D., Semrau, S., van Oudenaarden, A. & Mikkelsen, T. S. Characterization of directed differentiation by high-throughput single-cell RNA-Seq. Preprint at bioRxiv https://doi.org/10.1101/003236(2014)

  16. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  PubMed  Google Scholar 

  17. Tang, W. et al. bayNorm: Bayesian gene expression recovery, imputation and normalisation for single cell RNA-sequencing data. Preprint at bioRxiv https://doi.org/10.1101/384586(2018).

  18. Marguerat, S. et al. Quantitative analysis of fission yeast transcriptomes and proteomes in proliferating and quiescent cells. Cell 151, 671–683 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Newman, J. R. S. et al. Single-cell proteomic analysis of S. cerevisiae reveals the architecture of biological noise. Nature 441, 840–846 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Bar-Even, A. et al. Noise in protein expression scales with natural protein abundance. Nat. Genet. 38, 636–643 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Rustici, G. et al. Periodic gene expression program of the fission yeast cell cycle. Nat. Genet. 36, 809–817 (2004).

    Article  CAS  PubMed  Google Scholar 

  22. Peng, X. et al. Identification of cell cycle-regulated genes in fission yeast. Mol. Biol. Cell 16, 1026–1042 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Oliva, A. et al. The cell cycle-regulated genes of Schizosaccharomyces pombe. PLoS Biol. 3, e225 (2005).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Marguerat, S. et al. The more the merrier: comparative analysis of microarray studies on cell cycle-regulated genes in fission yeast. Yeast 23, 261–277 (2006).

    Article  CAS  PubMed  Google Scholar 

  25. Cooper, S. On a heuristic point of view concerning the expression of numerous genes during the cell cycle. IUBMB Life 64, 10–17 (2012).

    Article  CAS  PubMed  Google Scholar 

  26. Duncan, C. D. S., Rodríguez-López, M., Ruis, P., Bähler, J. & Mata, J. General amino acid control in fission yeast is regulated by a nonconserved transcription factor, with functions analogous to Gcn4/Atf4. Proc. Natl Acad. Sci. USA 115, E1829–E1838 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ravarani, C. N. J., Chalancon, G., Breker, M., de Groot, N. S. & Babu, M. M. Affinity and competition for TBP are molecular determinants of gene expression noise. Nat. Commun. 7, 10417 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Tirosh, I., Weinberger, A., Carmi, M. & Barkai, N. A genetic signature of interspecies variations in gene expression. Nat. Genet. 38, 830–834 (2006).

    Article  CAS  PubMed  Google Scholar 

  29. Landry, C. R., Lemos, B., Rifkin, S. A., Dickinson, W. J. & Hartl, D. L. Genetic properties influencing the evolvability of gene expression. Science 317, 118–121 (2007).

    Article  CAS  PubMed  Google Scholar 

  30. Lehner, B. Selection to minimise noise in living systems and its implications for the evolution of gene expression. Mol. Syst. Biol. 4, 170 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  31. Blake, W. J., KÆrn, M., Cantor, C. R. & Collins, J. J. Noise in eukaryotic gene expression. Nature 422, 633–637 (2003).

    Article  CAS  PubMed  Google Scholar 

  32. Weinberger, L. et al. Expression noise and acetylation profiles distinguish HDAC functions. Mol. Cell 47, 193–202 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Chen, D. et al. Global transcriptional responses of fission yeast to environmental stress. Mol. Biol. Cell 14, 214–229 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Pancaldi, V., Schubert, F. & Bähler, J. Meta-analysis of genome regulation and expression variability across hundreds of environmental and genetic perturbations in fission yeast. Mol. Biosyst. 6, 543–552 (2010).

    Article  CAS  PubMed  Google Scholar 

  35. Koch, E. N. et al. Conserved rules govern genetic interaction degree across species. Genome Biol. 13, R57 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  36. López-Maury, L., Marguerat, S. & Bähler, J. Tuning gene expression to changing environments: from rapid responses to evolutionary adaptation. Nat. Rev. Genet. 9, 583–593 (2008).

    Article  PubMed  Google Scholar 

  37. Rhind, N. et al. Comparative functional genomics of the fission yeasts. Science 332, 930–936 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Lane, K. et al. Measuring signaling and RNA-Seq in the same cell links gene expression to dynamic patterns of NF-κB activation. Cell Syst. 4, 458–469.e5 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cadwell, C. R. et al. Electrophysiological, transcriptomic and morphologic profiling of single neurons using Patch-seq. Nat. Biotechnol. 34, 199–203 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Nichterwitz, S. et al. Laser capture microscopy coupled with Smart-seq2 for precise spatial transcriptomic profiling. Nat. Commun. 7, 12139 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Turner, J. J., Ewald, J. C. & Skotheim, J. M. Cell size control in yeast. Curr. Biol. 22, R350–R359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kuang, Z. et al. High-temporal-resolution view of transcription and chromatin states across distinct metabolic states in budding yeast. Nat. Struct. Mol. Biol. 21, 854–863 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Silverman, S. J. et al. Metabolic cycling in single yeast cells from unsynchronized steady-state populations limited on glucose or phosphate. Proc. Natl Acad. Sci. USA 107, 6946–6951 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Slavov, N., Airoldi, E. M., van Oudenaarden, A. & Botstein, D. A conserved cell growth cycle can account for the environmental stress responses of divergent eukaryotes. Mol. Biol. Cell 23, 1986–1997 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Marguerat, S. & Bähler, J. Coordinating genome expression with cell size. Trends Genet. 28, 560–565 (2012).

    Article  CAS  PubMed  Google Scholar 

  46. Schmoller, K. M. & Skotheim, J. M. The biosynthetic basis of cell size control. Trends Cell Biol. 25, 793–802 (2015).

  47. Mata, J., Lyne, R., Burns, G. & Bähler, J. The transcriptional program of meiosis and sporulation in fission yeast. Nat. Genet. 32, 143–147 (2002).

    Article  CAS  PubMed  Google Scholar 

  48. Metzl-Raz, E. et al. Principles of cellular resource allocation revealed by condition-dependent proteome profiling. eLife 6, e28034 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Malecki, M. et al. Functional and regulatory profiling of energy metabolism in fission yeast. Genome Biol. 17, 240 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Moreno, S., Klar, A. & Nurse, P. Molecular genetic analysis of fission yeast Schizosaccharomyces pombe. Methods Enzymol. 194, 795–823 (1991).

    Article  CAS  PubMed  Google Scholar 

  51. Takahashi, C. N., Miller, A. W., Ekness, F., Dunham, M. J. & Klavins, E. A low cost, customizable turbidostat for use in synthetic circuit characterization. ACS Synth. Biol. 4, 32–38 (2015).

    Article  CAS  PubMed  Google Scholar 

  52. 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 

  53. Keifenheim, D. et al. Size-dependent expression of the mitotic activator Cdc25 suggests a mechanism of size control in fission yeast. Curr. Biol. 27, 1491–1497 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Smith, T., Heger, A. & Sudbery, I. UMI-tools: modeling sequencing errors in Unique Molecular Identifiers to improve quantification accuracy. Genome Res. 27, 491–499 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  56. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  57. Lun, A. T. L., Calero-Nieto, F. J., Haim-Vilmovsky, L., Göttgens, B. & Marioni, J. C. Assessing the reliability of spike-in normalization for analyses of single-cell RNA sequencing data. Genome Res. 27, 1795–1806 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Svensson, V. et al. Power analysis of single-cell RNA-sequencing experiments. Nat. Methods 14, 381–387 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Vallejos, C. A., Risso, D., Scialdone, A., Dudoit, S. & Marioni, J. C. Normalizing single-cell RNA sequencing data: challenges and opportunities. Nat. Methods 14, 565–571 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. 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 

  61. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bacher, R. et al. SCnorm: robust normalization of single-cell RNA-seq data. Nat. Methods 14, 584–586 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Lun, L., 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  Google Scholar 

  64. Lun, A. T. L., McCarthy, D. J. & Marioni, J. C. A step-by-step workflow for low-level analysis of single-cell RNA-seq data with Bioconductor. F1000Res. 5, 2122 (2016).

    PubMed  PubMed Central  Google Scholar 

  65. Ziegenhain, C., Vieth, B., Parekh, S., Hellmann, I. & Enard, W. Quantitative single-cell transcriptomics. Brief. Funct. Genomics 17, 220–232 (2018).

  66. 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 

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

    Article  PubMed  Google Scholar 

  68. Chen, H.-I. H., Jin, Y., Huang, Y. & Chen, Y. Detection of high variability in gene expression from single-cell RNA-seq profiling. BMC Genomics 17, 508 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  69. Jaakkola, M. K., Seyednasrollah, F., Mehmood, A. & Elo, L. L. Comparison of methods to detect differentially expressed genes between single-cell populations. Brief. Bioinform. 18, 735–743 (2017).

    PubMed  Google Scholar 

  70. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Breiman, L. Random forests. Mach. Learn. 45, 5–32 (2001).

    Article  Google Scholar 

  72. Bailey, T. L. & Machanick, P. Inferring direct DNA binding from ChIP-seq. Nucleic Acids Res. 40, e128 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. Livermore for his help during the initial part of this project and C. Stefanelli for her contribution to the development of bayNorm. We are grateful to S. Parrinello, A. Martinez-Segura and M. Priestman for their input on the manuscript. This research was supported by the UK Medical Research Council, a Leverhulme Research Project Grant (grant no. RPG-2014-408) and a Wellcome Trust Senior Investigator Award to J.B. (grant no. 095598/Z/11/Z). We used the computing resources of the UK Medical Bioinformatics partnership (UK MED-BIO), which is supported by the UK Medical Research Council (grant no. MR/L01632X/1) and the Imperial College High Performance Computing Service.

Author information

Authors and Affiliations

Authors

Contributions

M.S. set up the scRNA-seq protocol in yeast cells, performed all sequencing experiments and part of the computational analysis. W.T. and F.B. developed bayNorm and performed most computational analyses together with S.M. and V.S. X.M.S. performed all smFISH and growth experiments. A.K. developed the first generation of the single-cell isolation and PCR amplification protocol under the supervision of S.M. and J.B. L.G. assisted with the developement of the scRNA-seq protocol. S.M., V.S. and J.B. supervised this study. S.M., V.S., J.B., W.T. and M.S. wrote the paper.

Corresponding authors

Correspondence to Vahid Shahrezaei or Samuel Marguerat.

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 Figures 1–6, Supplementary References.

Reporting Summary

Supplementary Tables 1–10.

Samples description and statistics, description of experimental conditions, description of cells and ctRNA, raw counts, filtered normalized counts, gene statistics and features, smFISH data, smFISH probes sequences and fluorochromes, primers used for single-cell RNA sequencing, sequence analysis of HGV promoters.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Saint, M., Bertaux, F., Tang, W. et al. Single-cell imaging and RNA sequencing reveal patterns of gene expression heterogeneity during fission yeast growth and adaptation. Nat Microbiol 4, 480–491 (2019). https://doi.org/10.1038/s41564-018-0330-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41564-018-0330-4

This article is cited by

Search

Quick links

Nature Briefing Microbiology

Sign up for the Nature Briefing: Microbiology newsletter — what matters in microbiology research, free to your inbox weekly.

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