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:

A symmetric toggle switch explains the onset of random X inactivation in different mammals

Abstract

Gene-regulatory networks control the establishment and maintenance of alternative gene-expression states during development. A particular challenge is the acquisition of opposing states by two copies of the same gene, as in the case of the long non-coding RNA Xist in mammals at the onset of random X-chromosome inactivation (XCI). The regulatory principles that lead to stable mono-allelic expression of Xist remain unknown. Here, we uncover the minimal regulatory network that can ensure female-specific and mono-alleleic upregulation of Xist, by combining mathematical modeling and experimental validation of central model predictions. We identify a symmetric toggle switch as the basis for random mono-allelic upregulation of Xist, which reproduces data from several mutant, aneuploid and polyploid mouse cell lines with various Xist expression patterns. Moreover, this toggle switch explains the diversity of strategies employed by different species at the onset of XCI. In addition to providing a unifying conceptual framework with which to explore XCI across mammals, our study sets the stage for identifying the molecular mechanisms needed to initiate random XCI.

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: Comparison of alternative model structures.
Fig. 2: The cXR–tXA model can recapitulate Xist patterns in male, female, aneuploid and polyploid cell lines.
Fig. 3: The cXR–tXA model reproduces transient bi-allelic expression in different species.
Fig. 4: Bi-allelic Xist upregulation is reversible.
Fig. 5: Predicted cis-acting feedback can be mediated by antisense transcription.
Fig. 6: Transcriptional interferences at the Xist–Tsix locus.
Fig. 7: Simulation of Xist and Tsix mutant cell lines.

Similar content being viewed by others

Data availability

Source data for Figs. 3c,d, 4b,c,f,g,i, 5d and 6c–f and Supplementary Figs. 1 and 3a,b,d are available with the paper online. Data, code and simulations used in this study are available at https://github.com/verenamutzel/XCI_model under the MIT license. All other data and the cell line TX1072dT generated for this study are available upon reasonable request.

Code availability

All code and simulations used in this study are available at https://github.com/verenamutzel/XCI_model under the MIT license.

References

  1. 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 

  2. Sado, T. & Sakaguchi, T. Species-specific differences in X chromosome inactivation in mammals. Reproduction 146, R131–R139 (2013).

    Article  CAS  Google Scholar 

  3. Okamoto, I. et al. Eutherian mammals use diverse strategies to initiate X-chromosome inactivation during development. Nature 472, 370–374 (2011).

    Article  CAS  Google Scholar 

  4. Mak, W. et al. Reactivation of the paternal X chromosome in early mouse embryos. Science 303, 666–669 (2004).

    Article  CAS  Google Scholar 

  5. Okamoto, I., Otte, A. P., Allis, C. D., Reinberg, D. & Heard, E. Epigenetic dynamics of imprinted X inactivation during early mouse development. Science 303, 644–649 (2004).

    Article  CAS  Google Scholar 

  6. Petropoulos, S. et al. Single-cell RNA-seq reveals lineage and X chromosome dynamics in human preimplantation embryos. Cell 165, 1012–1026 (2016).

    Article  CAS  Google Scholar 

  7. Lee, J. T. & Lu, N. Targeted mutagenesis of Tsix leads to nonrandom X inactivation. Cell 99, 47–57 (1999).

    Article  CAS  Google Scholar 

  8. Migeon, B. R., Lee, C. H., Chowdhury, A. K. & Carpenter, H. Species differences in TSIX/Tsix reveal the roles of these genes in X-chromosome inactivation. Am. J. Hum. Genet. 71, 286–293 (2002).

    Article  CAS  Google Scholar 

  9. Vallot, C. et al. XACT noncoding RNA competes with XIST in the control of X chromosome activity during human early development. Cell Stem Cell 20, 102–111 (2017).

    Article  CAS  Google Scholar 

  10. Brown, C. J. et al. The human XIST gene: analysis of a 17 kb inactive X-specific RNA that contains conserved repeats and is highly localized within the nucleus. Cell 71, 527–542 (1992).

    Article  CAS  Google Scholar 

  11. Monkhorst, K. et al. The probability to initiate X chromosome inactivation is determined by the X to autosomal ratio and X chromosome specific allelic properties. PLoS ONE 4, e5616 (2009).

    Article  Google Scholar 

  12. Nora, E. P. et al. Spatial partitioning of the regulatory landscape of the X-inactivation centre. Nature 485, 381–385 (2012).

    Article  CAS  Google Scholar 

  13. Tian, D., Sun, S. & Lee, J. T. The long noncoding RNA, Jpx, is a molecular switch for X chromosome inactivation. Cell 143, 390–403 (2010).

    Article  CAS  Google Scholar 

  14. Furlan, G. et al. The Ftx noncoding locus controls x chromosome inactivation independently of its RNA products. Mol. Cell 70, 462–472.e8 (2018).

    Article  CAS  Google Scholar 

  15. Jonkers, I. et al. RNF12 is an X-encoded dose-dependent activator of X chromosome inactivation. Cell 139, 999–1011 (2009).

    Article  CAS  Google Scholar 

  16. Monkhorst, K., Jonkers, I., Rentmeester, E., Grosveld, F. & Gribnau, J. X. Inactivation counting and choice is a stochastic process: evidence for involvement of an X-linked activator. Cell 132, 410–421 (2008).

    Article  CAS  Google Scholar 

  17. Sun, S. et al. Jpx RNA activates Xist by evicting CTCF. Cell 153, 1537–1551 (2013).

    Article  CAS  Google Scholar 

  18. Endo, S., Takagi, N. & Sasaki, M. The late‐replicating X chromosome in digynous mouse triploid embryos. Dev. Genet. 3, 165–176 (1982).

    Article  Google Scholar 

  19. Henery, C. C., Bard, J. B. & Kaufman, M. H. Tetraploidy in mice, embryonic cell number, and the grain of the developmental map. Dev. Biol. 152, 233–241 (1992).

    Article  CAS  Google Scholar 

  20. Guyochin, A. et al. Live cell imaging of the nascent inactive X chromosome during the early differentiation process of naive ES cells towards epiblast stem cells. PLoS ONE 9, e116109 (2014).

    Article  Google Scholar 

  21. Sousa, E. J. et al. Exit from naive pluripotency induces a transient X chromosome inactivation-like state in males. Cell Stem Cell 22, 919–928.e6 (2018).

    Article  CAS  Google Scholar 

  22. Schulz, E. G. et al. The two active X chromosomes in female ESCs block exit from the pluripotent state by modulating the ESC signaling network. Cell Stem Cell 14, 203–216 (2014).

    Article  CAS  Google Scholar 

  23. Willard, H. F. & Carrel, L. Making sense (and antisense) of the X inactivation center. Proc. Natl Acad. Sci. USA 98, 10025–10027 (2001).

    Article  CAS  Google Scholar 

  24. Loos, F. et al. Xist and tsix transcription dynamics is regulated by the X-to-autosome ratio and semistable transcriptional states. Mol. Cell. Biol. 36, 2656–2667 (2016).

    Article  CAS  Google Scholar 

  25. Shearwin, K. E., Callen, B. P. & Egan, J. B. Transcriptional interference—a crash course. Trends Genet. 21, 339–345 (2005).

    Article  CAS  Google Scholar 

  26. Sneppen, K. et al. A mathematical model for transcriptional interference by RNA polymerase traffic in Escherichia coli. J. Mol. Biol. 346, 399–409 (2005).

    Article  CAS  Google Scholar 

  27. Nakanishi, H., Mitarai, N. & Sneppen, K. Dynamical analysis on gene activity in the presence of repressors and an interfering promoter. Biophys. J. 95, 4228–4240 (2008).

    Article  CAS  Google Scholar 

  28. Hobson, D. J., Wei, W., Steinmetz, L. M. & Svejstrup, J. Q. RNA polymerase II collision interrupts convergent transcription. Mol. Cell 48, 365–374 (2012).

  29. Wutz, A., Rasmussen, T. P. & Jaenisch, R. Chromosomal silencing and localization are mediated by different domains of Xist RNA. Nat. Genet. 30, 167–174 (2002).

    Article  CAS  Google Scholar 

  30. Penny, G. D., Kay, G. F., Sheardown, S. A., Rastan, S. & Brockdorff, N. Requirement for Xist in X chromosome inactivation. Nature 379, 131–137 (1996).

    Article  CAS  Google Scholar 

  31. Lee, J. T. Regulation of X-chromosome counting by Tsix and Xite sequences. Science 309, 768–771 (2005).

    Article  CAS  Google Scholar 

  32. Zhou, J. X. & Huang, S. Understanding gene circuits at cell-fate branch points for rational cell reprogramming. Trends Genet. 27, 55–62 (2011).

    Article  CAS  Google Scholar 

  33. Lee, J. T., Davidow, L. S. & Warshawsky, D. Tsix, a gene antisense to Xist at the X-inactivation centre. Nat. Genet. 21, 400–404 (1999).

    Article  CAS  Google Scholar 

  34. Navarro, P. et al. Molecular coupling of Xist regulation and pluripotency. Science 321, 1693–1695 (2008).

    Article  CAS  Google Scholar 

  35. Donohoe, M. E., Silva, S. S., Pinter, S. F., Xu, N. & Lee, J. T. The pluripotency factor Oct4 interacts with Ctcf and also controls X-chromosome pairing and counting. Nature 460, 128–132 (2009).

    Article  CAS  Google Scholar 

  36. Schulz, E. G. & Heard, E. Role and control of X chromosome dosage in mammalian development. Curr. Opin. Genet. Dev. 23, 109–115 (2013).

    Article  CAS  Google Scholar 

  37. Gontan, C. et al. RNF12 initiates X-chromosome inactivation by targeting REX1 for degradation. Nature 485, 386–390 (2012).

    Article  CAS  Google Scholar 

  38. Gontan, C. et al. REX1 is the critical target of RNF12 in imprinted X chromosome inactivation in mice. Nat. Comm. 9, 4752 (2018).

    Article  Google Scholar 

  39. Barakat, T. S. et al. RNF12 activates Xist and is essential for X chromosome inactivation. PLoS Genet. 7, e1002001 (2011).

    Article  CAS  Google Scholar 

  40. Wang, F. et al. Rlim-dependent and -independent pathways for X chromosome inactivation in female ESCs. Cell Rep. 21, 3691–3699 (2017).

    Article  CAS  Google Scholar 

  41. Shin, J. et al. RLIM is dispensable for X-chromosome inactivation in the mouse embryonic epiblast. Nature 511, 86–89 (2014).

    Article  CAS  Google Scholar 

  42. Quinn, J. J. & Chang, H. Y. Unique features of long non-coding RNA biogenesis and function. Nat. Rev. Genet. 17, 47–62 (2016).

    Article  CAS  Google Scholar 

  43. Navarro, P., Page, D. R., Avner, P. & Rougeulle, C. Tsix-mediated epigenetic switch of a CTCF-flanked region of the Xist promoter determines the Xist transcription program. Genes Dev. 20, 2787–2792 (2006).

    Article  CAS  Google Scholar 

  44. Sado, T., Hoki, Y. & Sasaki, H. Tsix silences Xist through modification of chromatin structure. Dev. Cell 9, 159–165 (2005).

    Article  CAS  Google Scholar 

  45. Khan, S. A., Audergon, P. N. C. B. & Payer, B. X-chromosome activity in naive human pluripotent stem cells—are we there yet? Stem Cell Invest. 4, 54–54 (2017).

    Article  Google Scholar 

  46. Rose, N. R. & Klose, R. J. Understanding the relationship between DNA methylation and histone lysine methylation. Biochim. Biophys. Acta 1839, 1362–1372 (2014).

    Article  CAS  Google Scholar 

  47. Inoue, A., Jiang, L., Lu, F., Suzuki, T. & Zhang, Y. Maternal H3K27me3 controls DNA methylation-independent imprinting. Nature 547, 419–424 (2017).

    Article  CAS  Google Scholar 

  48. Angel, A., Song, J., Dean, C. & Howard, M. A Polycomb-based switch underlying quantitative epigenetic memory. Nature 476, 105–108 (2011).

    Article  CAS  Google Scholar 

  49. Dodd, I. B., Micheelsen, M. A., Sneppen, K. & Thon, G. Theoretical analysis of epigenetic cell memory by nucleosome modification. Cell 129, 813–822 (2007).

    Article  CAS  Google Scholar 

  50. Yang, H., Howard, M. & Dean, C. Antagonistic roles for H3K36me3 and H3K27me3 in the cold-induced epigenetic switch at arabidopsis FLC. Curr. Biol. 24, 1793–1797 (2014).

    Article  CAS  Google Scholar 

  51. Sakata, Y. et al. Defects in dosage compensation impact global gene regulation in the mouse trophoblast. Development 144, 2784–2797 (2017).

    Article  CAS  Google Scholar 

  52. Barakat, T. S. et al. The trans-activator RNF12 and cis-acting elements effectuate X chromosome inactivation independent of X-pairing. Mol. Cell 53, 965–978 (2014).

    Article  CAS  Google Scholar 

  53. Gillespie, D. T. Exact stochastic simulation of coupled chemical reactions. J. Phys. Chem. 81, 2340–2361 (1977).

    Article  CAS  Google Scholar 

  54. Jonkers, I., Kwak, H., Lis, J. T. & Struhl, K. Genome-wide dynamics of Pol II elongation and its interplay with promoter proximal pausing, chromatin, and exons. eLife 3, e02407 (2014).

    Article  Google Scholar 

  55. Sun, B. K., Deaton, A. M. & Lee, J. T. A transient heterochromatic state in Xist preempts X inactivation choice without RNA stabilization. Mol. Cell 21, 617–628 (2006).

    Article  CAS  Google Scholar 

  56. Yamada, N. et al. Xist exon 7 contributes to the stable localization of Xist RNA on the inactive X-chromosome. PLoS Genet. 11, e1005430 (2015).

    Article  Google Scholar 

  57. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  Google Scholar 

  58. Chaumeil, J., Augui, S., Chow, J. C. & Heard, E. Combined immunofluorescence, RNA fluorescent in situ hybridization, and DNA fluorescent in situ hybridization to study chromatin changes, transcriptional activity, nuclear organization, and X-chromosome inactivation. Methods Mol. Biol. 463, 297–308 (2008).

    Article  CAS  Google Scholar 

  59. Giorgetti, L. et al. Predictive polymer modeling reveals coupled fluctuations in chromosome conformation and transcription. Cell 157, 950–963 (2014).

    Article  CAS  Google Scholar 

  60. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2012).

    Article  Google Scholar 

  61. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Wutz (ETH Zürich, Switzerland) for the TXY (Xist-tetOP) and TXY∆A (Xist-∆SX-tetOP) mESC lines. We thank the staff of the Max Planck Institute for Molecular Genetics (MPIMG) and PICTIBiSA@BDD imaging facilities for technical assistance, the MPIMG sequencing core facility of sequencing services and the MPIMG IT facility for support in using the computing cluster. We thank R. Galupa for valuable feedback on the manuscript. This work was funded by a Human Frontier Science Program (HFSP) long-term fellowship (LT000597/2010-L) to E.G.S., a Grant-in-Aid for Specially Promoted Research from the Japan Society for the Promotion of Science (JSPS) (17H06098) to M.S., and by a Japan Science and Technology Agency (JST) Exploratory Research for Advanced Technology (ERATO) grant (JPMJER1104) to M.S., I.O. and M.S., and JSPS KAKENHI grants (25291076 and 18K06030) to I.O. Reseach in the laboratory of E.G.S. is funded by the Max-Planck Research Group Leader program and and the German Ministry of Science and Education (BMBF) through the grant E:bio Module III - Xnet. V.M. is supported by the DFG (GRK1772, Computational Systems Biology). Research in the laboratory of L.G. is funded by an ERC Starting grant (759366).

Author information

Authors and Affiliations

Authors

Contributions

E.G.S., E.H. and I.O. conceived the study. V.M. and E.G.S. wrote scripts and performed simulations. V.M., I.O., I.D., L.G. and E.G.S. carried out the experiments. E.G.S. and V.M. wrote the paper with input from E.H. and L.G. E.G.S., E.H. and M.S. supervised the study. E.G.S., E.H., L.G., I.O. and M.S. acquired funding.

Corresponding author

Correspondence to Edda G. Schulz.

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.

Integrated supplementary information

Supplementary Figure 1 The cXR-tXA model can reproduce up-regulation of Xist in differentiating mESCs.

Fraction of cells exhibiting mono-allelic (light grey) and bi-allelic Xist expression (dark grey) during differentiation of mESC line TX1072. Experimental data (circles) is shown together with a simulation using the parameter set that best explains the data. The number of cells analyzed is given on top. The data was pooled from 3 independent experiments.

Source data

Supplementary Figure 2 Transient bi-allelic up-regulation of Xist in the cXR-tXA model.

(a) For all parameter sets that reproduced mono-allelic Xist up-regulation in the cXR-tXA model, the maximal fraction of cells with bi-allelic Xist expression observed during the simulation is shown as a function of the the ratio of switch-ON time (first time point, when Xist levels reach 20% of the high steady state) and tXA silencing delay (siltXA). If Xist up-regulation is slow (high Switch-ON time), it will normally occur one allele at a time. Subsequent silencing will shift the system to the bistable regime (cp. Fig. 2e) and thereby lock in the mono-allelic state before Xist up-regulation from the other X chromosome occurs. This results in a low frequency of bi-allelically expressing cells as observed in mice. If Xist up-regulation is rapid and silencing is slow (long silencing delay siltXA), Xist will initially be expressed from two alleles as observed in rabbit embryos. In this scenario the choice of the inactive X can subsequently occur through mono-allelic silencing of tXA and cXR. Alternatively, silencing of both alleles might reverse Xist up-regulation completely as Xist expression is unstable if both tXA alleles are silenced such that the cell can undertake a second attempt to reach the mono-allelic state. (b) Simulation of bi-allelic expression upon reduced Xist-mediated silencing as observed in human embryos, assuming that in the first 4 days of the simulation either silencing and cXR expression is absent (left) or that cXR is silenced partially (dampening), while tXA is unaffected by Xist (right). Boxplots show the percentage of mono- and bi-allelically expressing cells for 100 randomly chosen parameter sets that can reproduce mono-allelic Xist up-regulation (center line, median; box limits, upper and lower quartiles; whiskers, most extreme data points not considered outliers; points, outliers).

Supplementary Figure 3 Bi-allelic up-regulation of Xist is reversible.

(a) Simulation of doxycycline treatment one day before the onset of differentiation (linked to Fig. 4a–c). Boxplots show the frequency of mono-allelic (left) and bi-allelic Xist expression (right) in dox-treated (grey) and control cells (black) for 100 parameter sets that could reproduce mono-allelic Xist up-regulation. (b) Boxplots show the simulation results for artificial bi-allelic Xist induction as described in Fig. 4e in the main text, using the same parameters sets as in (a). On each box, the central mark indicates the median, and the bottom and top edges of the box indicate the 25th and 75th percentiles, respectively. The whiskers extend to the most extreme data points not considered outliers, and the outliers are plotted individually. (c, d) Bi-allelic Xist up-regulation is artificially induced by treating TX1072dT cells with doxycycline after 48h of differentiation (cp. Fig. 4e). Cells were treated with EdU to assay proliferation through measuring its incorporation into DNA during replication. EdU was labeled fluorescently through Click-it chemistry and Xist was visualized by RNA FISH. (c) The EdU-positive fraction was quantified at the indicated time points within cells expressing Xist mono- (black) and bi-allelically (grey). Mean and s.d. of n=3 independent experiments are shown, in each replicate at least 50 cells were counted per group, except for bi-allelic cells at 48h. * paired two-sample two-sided T-test. Scale bar indicates 5 μm.

Source data

Supplementary Figure 4 Transcriptional interference can generate a precise threshold, which is required for reliable mono-allelic Xist up-regulation.

(a) Steady state Xist levels simulated deterministically (see Fig. 2e) to indicate that a sharp threshold is required between a single (1x) and a double (2x) tXA dose. (b) Maintenance of the XaXi state was simulated by initiating an allele either from the Xa (light green) or from the Xi state (dark green). For an example parameter set (kT=113 h-1, t1/2repr =0.7 h) mean and standard deviation of Xist expression across 100 cells from the chromosomes that initiated as Xa (light green) and Xi (dark green), respectively, is shown for different values of kX for the full Xist-Tsix model (left) and the reduced model without transcriptional interference (right). The vertical lines indicate the kX threshold value, above which >1 (Thlow, red) or >99 (Thhigh, grey) out of 100 cells up-regulate Xist from the Xa. (c) Distribution of the Thhigh-to-Thlow ratio (red and grey in (b)) across all parameter sets of the Full model (grey) and the reduced model without transcriptional interference (black). Since tXA reduces kX 2-fold upon Xist up-regualtion a threshold ratio of <2 is required to allow reliable Xist up-regulation with a double dose (2x) of tXA and stable maintenance of the XaXi state with a single dose (1x). This is only possible in the Full model with transcriptional interference.

Supplementary Figure 5 Transcriptional interference at the XistTsix locus.

(a–f) TXY and TXY∆A ESCs were treated with doxycycline for 24 hours and nascent transcription of Xist and Tsix (5′ and 3′) was assessed by RNA FISH. (ac) Quantification of 3 biological replicates, where each dot represents the measured signal intensities of a single allele. Grey lines indicate the detection threshold estimated from negative control regions. (df) Box plots of Tsix signal intensity at Xist+ (green) and Xist- alleles (black) in the two cell lines as indicated for the data shown in (a-c); dotted lines indicate the detection threshold (center line, median; box limits, upper and lower quartiles; whiskers, most extreme data points not considered outliers; points, outliers).

Supplementary Figure 6 Also a reduced overlap between Xist and Tsix as in the human locus allows mono-allelic up-regulation of Xist.

(a, c) Schematic representation of the Xist-Tsix locus architecture in the mouse (a) and the human (c) genome, respectively. (b, d) Distribution of the mean frequency of mono-allelic Xist up-regulation across all parameter sets tested, in simulations assuming the locus architecture shown in (a) and (c), respectively. For details see Supplementary Note 1 (section 3.5). (e, f) Simulation of Xist up-regulation using the model with the human locus architecture in (c) for one example parameter set, showing three individual cells (e) and a population of 100 cells (f). Light and dark green in (e) represent Xist levels expressed from the two X chromosomes, light and dark grey in (f) represent mono- and bi-allelic Xist expression, as indicated.

Supplementary information

Supplementary Information

Supplementary Figure 1–6 and Supplementary Notes 1–3

Reporting Summary

Supplementary Table 1

Reagent Summary. Detailed information on all qPCR and pyrosequencing primers, antibodies, Stellaris FISH probes and amplicons assessed by amplicon sequencing.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mutzel, V., Okamoto, I., Dunkel, I. et al. A symmetric toggle switch explains the onset of random X inactivation in different mammals. Nat Struct Mol Biol 26, 350–360 (2019). https://doi.org/10.1038/s41594-019-0214-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41594-019-0214-1

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