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:

X-chromosome upregulation is driven by increased burst frequency

Abstract

Ohno's hypothesis postulates that upregulation of X-linked genes rectifies their dosage imbalance relative to autosomal genes, which are present in two active copies per cell. Here we have dissected X-chromosome upregulation into the kinetics of transcription, inferred from allele-specific single-cell RNA sequencing data from somatic and embryonic mouse cells. We confirmed increased X-chromosome expression levels in female and male cells and found that the X chromosome achieved upregulation by elevated burst frequencies. By monitoring transcriptional kinetics in differentiating female mouse embryonic stem cells, we found that increased burst frequency was established on the active X chromosome when X inactivation took place on the other allele. Thus, our study provides mechanistic insights into X-chromosome upregulation.

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: Investigating X-chromosome upregulation by transcriptional kinetics.
Fig. 2: Classification of cells based on X inactivation status.
Fig. 3: Elevated expression levels of X-linked genes.
Fig. 4: Transcriptional kinetics of X-chromosome upregulation.
Fig. 5: X-chromosome burst frequencies during differentiation of ESCs.

Similar content being viewed by others

Data availability

The sequencing data are available at the European Nucleotide Archive (E-MTAB-7098), ArrayExpress (E-MTAB-6385) and GEO (GSE74155).

Code availability

The computational code used for calculations and plotting of data is available at https://sourceforge.net/projects/kinetics-of-x-upregulation.

References

  1. Berta, P. et al. Genetic-evidence equating SRY and the testis-determining factor. Nature 348, 448–450 (1990).

    Article  CAS  Google Scholar 

  2. Wallis, M. C., Waters, P. D. & Graves, J. A. M. Sex determination in mammals—before and after the evolution of SRY. Cell. Mol. Life Sci. 65, 3182–3195 (2008).

    Article  CAS  Google Scholar 

  3. Charlesworth, B. The evolution of chromosomal sex determination and dosage compensation. Curr. Biol. 6, 149–162 (1996).

    Article  CAS  Google Scholar 

  4. Ohno, S. Sex Chromosomes and Sex-linked Genes (Springer, 1967).

  5. Lyon, M. F. Gene action in X-chromosome of the mouse (Mus musculus L.). Nature 190, 372–373 (1961).

    Article  CAS  Google Scholar 

  6. Gupta, V. et al. Global analysis of X-chromosome dosage compensation. J. Biol. 5, 3 (2006).

    Article  Google Scholar 

  7. Nguyen, D. K. & Disteche, C. M. Dosage compensation of the active X chromosome in mammals. Nat. Genet. 38, 47–53 (2006).

    Article  CAS  Google Scholar 

  8. Lin, H. et al. Dosage compensation in the mouse balances up-regulation and silencing of X-linked genes. PLoS Biol. 5, 2809–2820 (2007).

    Article  CAS  Google Scholar 

  9. Lin, H. et al. Relative overexpression of X-linked genes in mouse embryonic stem cells is consistent with Ohno's hypothesis. Nat. Genet. 43, 1169–1170 (2011); author reply 43, 1171–1172.

  10. Deng, X. et al. Evidence for compensatory upregulation of expressed X-linked genes in mammals, Caenorhabditis elegans and Drosophila melanogaster. Nat. Genet. 43, 1179–1185 (2011).

    Article  CAS  Google Scholar 

  11. Julien, P. et al. Mechanisms and evolutionary patterns of mammalian and avian dosage compensation. PLoS Biol. 10, e1001328 (2012).

    Article  CAS  Google Scholar 

  12. Xiong, Y. et al. RNA sequencing shows no dosage compensation of the active X-chromosome. Nat. Genet. 42, 1043–1047 (2010).

    Article  CAS  Google Scholar 

  13. Wang, M., Lin, F., Xing, K. & Liu, L. Random X-chromosome inactivation dynamics in vivo by single-cell RNA sequencing. BMC Genom. 18, 90 (2017).

    Article  Google Scholar 

  14. Lin, F. Q., Xing, K., Zhang, J. Z. & He, X. L. Expression reduction in mammalian X chromosome evolution refutes Ohno's hypothesis of dosage compensation. Proc. Natl Acad. Sci. USA 109, 11752–11757 (2012).

    Article  CAS  Google Scholar 

  15. Li, X. et al. Dosage compensation in the process of inactivation/reactivation during both germ cell development and early embryogenesis in mouse. Sci. Rep. 7, 3729 (2017).

    Article  Google Scholar 

  16. Sangrithi, M. N. et al. Non-canonical and sexually dimorphic X dosage compensation states in the mouse and human germline. Dev. Cell 40, 289–301.e3 (2017).

    Article  CAS  Google Scholar 

  17. Yang, J.R. et al. Dosage sensitivity of X-linked genes in human embryonic single cells. BMC Genom. 20, 42 (2019).

    Article  Google Scholar 

  18. Kivioja, T. et al. Counting absolute numbers of molecules using unique molecular identifiers. Nat. Methods 9, 72–U183 (2012).

    Article  CAS  Google Scholar 

  19. Ziegenhain, C. et al. Comparative analysis of single-cell RNA sequencing methods. Mol. Cell 65, 631–643.e4 (2017).

    Article  CAS  Google Scholar 

  20. Levine, M. & Tjian, R. Transcription regulation and animal diversity. Nature 424, 147–151 (2003).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  22. Larsson, A. J. M. et al. Genomic encoding of transcriptional burst kinetics. Nature 565, 251–254 (2019).

    Article  CAS  Google Scholar 

  23. Symmons, O. et al. Allele-specific RNA imaging shows that allelic imbalances can arise in tissues through transcriptional bursting. PLoS Genet. 15, e1007874 (2019).

    Article  Google Scholar 

  24. Peccoud, J. & Ycart, B. Markovian modeling of gene-product synthesis. Theor. Popul. Biol. 48, 222–234 (1995).

    Article  Google Scholar 

  25. Mank, J. E. Sex chromosome dosage compensation: definitely not for everyone. Trends Genet. 29, 677–683 (2013).

    Article  CAS  Google Scholar 

  26. Deng, X. et al. Mammalian X upregulation is associated with enhanced transcription initiation, RNA half-life and MOF-mediated H4K16 acetylation. Dev. Cell 25, 55–68 (2013).

    Article  CAS  Google Scholar 

  27. Faucillion, M. L. & Larsson, J. Increased expression of X-linked genes in mammals is associated with a higher stability of transcripts and an increased ribosome density. Genome Biol. Evol. 7, 1039–1052 (2015).

    Article  CAS  Google Scholar 

  28. Augui, S., Nora, E. P. & Heard, E. Regulation of X-chromosome inactivation by the X-inactivation centre. Nat. Rev. Genet. 12, 429–442 (2011).

    Article  CAS  Google Scholar 

  29. Wutz, A. Gene silencing in X-chromosome inactivation: advances in understanding facultative heterochromatin formation. Nat. Rev. Genet. 12, 542–553 (2011).

    Article  CAS  Google Scholar 

  30. Chen, G. et al. Single-cell analyses of X chromosome inactivation dynamics and pluripotency during differentiation. Genome Res. 26, 1342–1354 (2016).

    Article  CAS  Google Scholar 

  31. Pintacuda, G. & Cerase, A. X inactivation lessons from differentiating mouse embryonic stem cells. Stem Cell Rev. Rep. 11, 699–705 (2015).

    Article  CAS  Google Scholar 

  32. Deng, X., Berletch, J. B., Nguyen, D. K. & Disteche, C. M. X-chromosome regulation: diverse patterns in development, tissues and disease. Nat. Rev. Genet. 15, 367–378 (2014).

    Article  CAS  Google Scholar 

  33. Yang, F., Babak, T., Shendure, J. & Disteche, C. M. Global survey of escape from X inactivation by RNA-sequencing in mouse. Genome Res. 20, 614–622 (2010).

    Article  CAS  Google Scholar 

  34. Yildirim, E., Sadreyev, R. I., Pinter, S. F. & Lee, J. T. X-chromosome hyperactivation in mammals via nonlinear relationships between chromatin states and transcription. Nat. Struct. Mol. Biol. 19, 56–U72 (2012).

    Article  CAS  Google Scholar 

  35. Bartman, C. R. et al. Transcriptional burst initiation and polymerase pause release are key control points of transcriptional regulation. Mol. Cell 73, 519–532.e4 (2019).

    Article  CAS  Google Scholar 

  36. Bartman, C. R., Hsu, S. C., Hsiung, C. C. S., Raj, A. & Blobel, G. A. Enhancer regulation of transcriptional bursting parameters revealed by forced chromatin looping. Mol. Cell 62, 237–247 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  39. Reinius, B. et al. Analysis of allelic expression patterns in clonal somatic cells by single-cell RNA-seq. Nat. Genet. 48, 1430–1435 (2016).

    Article  CAS  Google Scholar 

  40. Herzog, V. A. et al. Thiol-linked alkylation of RNA to assess expression dynamics. Nat. Methods 14, 1198–1204 (2017).

    Article  CAS  Google Scholar 

  41. Li, B. et al. A comprehensive mouse transcriptomic BodyMap across 17 tissues by RNA-seq. Sci. Rep. 7, 4200 (2017).

    Article  Google Scholar 

  42. Chadwick, L. H., Pertz, L. M., Broman, K. W., Bartolomei, M. S. & Willard, H. F. Genetic control of X chromosome inactivation in mice: definition of the Xce candidate interval. Genetics 173, 2103–2110 (2006).

    Article  CAS  Google Scholar 

  43. Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).

    Article  Google Scholar 

Download references

Acknowledgements

This study was made possible by grants to B.R. from the Ragnar Söderberg Foundation, the Swedish Research Council (2017-01723) and Åke Wiberg’s Foundation; and grants to R.S. from the European Research Council (648842) and the Swedish Research Council (2017-01062). The authors thank D. Camsund for providing insightful comments to the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

B.R. conceived and supervised the study. A.J.M.L. and B.R. analyzed the data and wrote the manuscript. A.J.M.L., C.C., R.S. and B.R. participated in interpreting the data and editing the manuscript.

Corresponding author

Correspondence to Björn Reinius.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Anke Sparmann was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Supplementary information

Supplementary Information

Supplementary Figs. 1–9 and Supplementary Tables 1 and 2.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Larsson, A.J.M., Coucoravas, C., Sandberg, R. et al. X-chromosome upregulation is driven by increased burst frequency. Nat Struct Mol Biol 26, 963–969 (2019). https://doi.org/10.1038/s41594-019-0306-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0306-y

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