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:

p53 pulses lead to distinct patterns of gene expression albeit similar DNA-binding dynamics

Abstract

The dynamics of transcription factors play important roles in a variety of biological systems. However, the mechanisms by which these dynamics are decoded into different transcriptional responses are not well understood. Here we focus on the dynamics of the tumor-suppressor protein p53, which exhibits a series of pulses in response to DNA damage. We performed time course RNA sequencing (RNA-seq) and chromatin immunoprecipitation sequencing (ChIP-seq) measurements to determine how p53 oscillations are linked with gene expression genome wide. We discovered multiple distinct patterns of gene expression in response to p53 pulses. Surprisingly, p53-binding dynamics were uniform across all genomic loci, even for genes that exhibited distinct mRNA dynamics. Using a mathematical model, supported by additional experimental measurements in response to sustained p53 input, we determined that p53 binds to and activates transcription of its target genes uniformly, whereas post-transcriptional mechanisms are responsible for the differences in gene expression dynamics.

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

Figure 1: Time course mRNA-seq reveals distinct clusters of gene expression dynamics in response to γ-irradiation.
Figure 2: Time course p53 ChIP-seq experiment shows pulsatile dynamics genome wide.
Figure 3: p53 target genes with distinct expression dynamics show similar dynamics of p53 binding.
Figure 4: Mathematical model links the input–output relationship between p53 protein dynamics and the dynamics of its target genes genome wide.
Figure 5: Mathematical model can predict gene expression for sustained p53 input dynamics.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Cai, L., Dalal, C.K. & Elowitz, M.B. Frequency-modulated nuclear localization bursts coordinate gene regulation. Nature 455, 485–490 (2008).

    Article  CAS  Google Scholar 

  2. Hoffmann, A., Levchenko, A., Scott, M.L. & Baltimore, D. The IkappaB-NF-kappaB signaling module: temporal control and selective gene activation. Science 298, 1241–1245 (2002).

    Article  CAS  Google Scholar 

  3. Purvis, J.E. et al. p53 dynamics control cell fate. Science 336, 1440–1444 (2012).

    Article  CAS  Google Scholar 

  4. Paek, A.L., Liu, J.C., Loewer, A., Forrester, W.C. & Lahav, G. Cell-to-cell variation in p53 dynamics leads to fractional killing. Cell 165, 631–642 (2016).

    Article  CAS  Google Scholar 

  5. Hao, N. & O'Shea, E.K. Signal-dependent dynamics of transcription factor translocation controls gene expression. Nat. Struct. Mol. Biol. 19, 31–39 (2011).

    Article  Google Scholar 

  6. Covert, M.W., Leung, T.H., Gaston, J.E. & Baltimore, D. Achieving stability of lipopolysaccharide-induced NF-kappaB activation. Science 309, 1854–1857 (2005).

    Article  CAS  Google Scholar 

  7. Batchelor, E., Loewer, A., Mock, C. & Lahav, G. Stimulus-dependent dynamics of p53 in single cells. Mol. Syst. Biol. 7, 488 (2011).

    Article  Google Scholar 

  8. Hansen, A.S. & O'Shea, E.K. Encoding four gene expression programs in the activation dynamics of a single transcription factor. Curr. Biol. 26, R269–R271 (2016).

    Article  CAS  Google Scholar 

  9. Werner, S.L., Barken, D. & Hoffmann, A. Stimulus specificity of gene expression programs determined by temporal control of IKK activity. Science 309, 1857–1861 (2005).

    Article  CAS  Google Scholar 

  10. Lahav, G. et al. Dynamics of the p53-Mdm2 feedback loop in individual cells. Nat. Genet. 36, 147–150 (2004).

    Article  CAS  Google Scholar 

  11. Lev Bar-Or, R. et al. Generation of oscillations by the p53-Mdm2 feedback loop: a theoretical and experimental study. Proc. Natl. Acad. Sci. USA 97, 11250–11255 (2000).

    Article  CAS  Google Scholar 

  12. Weinberg, R.L., Veprintsev, D.B., Bycroft, M. & Fersht, A.R. Comparative binding of p53 to its promoter and DNA recognition elements. J. Mol. Biol. 348, 589–596 (2005).

    Article  CAS  Google Scholar 

  13. Qian, H., Wang, T., Naumovski, L., Lopez, C.D. & Brachmann, R.K. Groups of p53 target genes involved in specific p53 downstream effects cluster into different classes of DNA binding sites. Oncogene 21, 7901–7911 (2002).

    Article  CAS  Google Scholar 

  14. Kracikova, M., Akiri, G., George, A., Sachidanandam, R. & Aaronson, S.A. A threshold mechanism mediates p53 cell fate decision between growth arrest and apoptosis. Cell Death Differ. 20, 576–588 (2013).

    Article  CAS  Google Scholar 

  15. Murray-Zmijewski, F., Slee, E.A. & Lu, X. A complex barcode underlies the heterogeneous response of p53 to stress. Nat. Rev. Mol. Cell Biol. 9, 702–712 (2008).

    Article  CAS  Google Scholar 

  16. Flores, E.R. et al. p63 and p73 are required for p53-dependent apoptosis in response to DNA damage. Nature 416, 560–564 (2002).

    Article  CAS  Google Scholar 

  17. Samuels-Lev, Y. et al. ASPP proteins specifically stimulate the apoptotic function of p53. Mol. Cell 8, 781–794 (2001).

    Article  CAS  Google Scholar 

  18. Oda, K. et al. p53AIP1, a potential mediator of p53-dependent apoptosis, and its regulation by Ser-46-phosphorylated p53. Cell 102, 849–862 (2000).

    Article  CAS  Google Scholar 

  19. Smeenk, L. et al. Role of p53 serine 46 in p53 target gene regulation. PLoS One 6, e17574 (2011).

    Article  CAS  Google Scholar 

  20. Sykes, S.M. et al. Acetylation of the p53 DNA-binding domain regulates apoptosis induction. Mol. Cell 24, 841–851 (2006).

    Article  CAS  Google Scholar 

  21. Nikulenkov, F. et al. Insights into p53 transcriptional function via genome-wide chromatin occupancy and gene expression analysis. Cell Death Differ. 19, 1992–2002 (2012).

    Article  CAS  Google Scholar 

  22. Verfaillie, A. et al. Multiplex enhancer-reporter assays uncover unsophisticated TP53 enhancer logic. Genome Res. 26, 882–895 (2016).

    Article  CAS  Google Scholar 

  23. Porter, J.R., Fisher, B.E. & Batchelor, E. p53 pulses diversify target gene expression dynamics in an mRNA half-life-dependent manner and delineate co-regulated target gene subnetworks. Cell Syst. 2, 272–282 (2016).

    Article  CAS  Google Scholar 

  24. Melanson, B.D. et al. The role of mRNA decay in p53-induced gene expression. RNA 17, 2222–2234 (2011).

    Article  CAS  Google Scholar 

  25. Batchelor, E., Mock, C.S., Bhan, I., Loewer, A. & Lahav, G. Recurrent initiation: a mechanism for triggering p53 pulses in response to DNA damage. Mol. Cell 30, 277–289 (2008).

    Article  CAS  Google Scholar 

  26. Wei, C.L. et al. A global map of p53 transcription-factor binding sites in the human genome. Cell 124, 207–219 (2006).

    Article  CAS  Google Scholar 

  27. El-Deiry, W.S. et al. WAF1, a potential mediator of p53 tumor suppression. Cell 75, 817–825 (1993).

    Article  CAS  Google Scholar 

  28. Botcheva, K., McCorkle, S.R., McCombie, W.R., Dunn, J.J. & Anderson, C.W. Distinct p53 genomic binding patterns in normal and cancer-derived human cells. Cell Cycle 10, 4237–4249 (2011).

    Article  CAS  Google Scholar 

  29. El-Deiry, W.S., Kern, S.E., Pietenpol, J.A., Kinzler, K.W. & Vogelstein, B. Definition of a consensus binding site for p53. Nat. Genet. 1, 45–49 (1992).

    Article  CAS  Google Scholar 

  30. Riley, T., Sontag, E., Chen, P. & Levine, A. Transcriptional control of human p53-regulated genes. Nat. Rev. Mol. Cell Biol. 9, 402–412 (2008).

    Article  CAS  Google Scholar 

  31. Fischer, M., Steiner, L. & Engeland, K. The transcription factor p53: not a repressor, solely an activator. Cell Cycle 13, 3037–3058 (2014).

    Article  CAS  Google Scholar 

  32. Schlereth, K. et al. Characterization of the p53 cistrome–DNA binding cooperativity dissects p53′s tumor suppressor functions. PLoS Genet. 9, e1003726 (2013).

    Article  CAS  Google Scholar 

  33. Rashi-Elkeles, S. et al. Parallel profiling of the transcriptome, cistrome, and epigenome in the cellular response to ionizing radiation. Sci. Signal. 7, rs3 (2014).

    Article  Google Scholar 

  34. Schueler, M. et al. Differential protein occupancy profiling of the mRNA transcriptome. Genome Biol. 15, R15 (2014).

    Article  Google Scholar 

  35. Zambrano, S., De Toma, I., Piffer, A., Bianchi, M.E. & Agresti, A. NF-κB oscillations translate into functionally related patterns of gene expression. eLife 5, e09100 (2016).

    Article  Google Scholar 

  36. Hao, S. & Baltimore, D. The stability of mRNA influences the temporal order of the induction of genes encoding inflammatory molecules. Nat. Immunol. 10, 281–288 (2009).

    Article  CAS  Google Scholar 

  37. Brummelkamp, T.R., Bernards, R. & Agami, R. A system for stable expression of short interfering RNAs in mammalian cells. Science 296, 550–553 (2002).

    Article  CAS  Google Scholar 

  38. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  Google Scholar 

  39. Kuleshov, M.V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  Google Scholar 

  40. Chen, E.Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).

    Article  Google Scholar 

  41. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S.L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  Google Scholar 

  42. Li, Q., Brown, J.B., Huang, H. & Bickel, P.J. Measuring reproducibility of high-throughput experiments. Ann. Appl. Stat. 5, 1752–1779 (2011).

    Article  Google Scholar 

  43. Landt, S.G. et al. ChIP-seq guidelines and practices of the ENCODE and modENCODE consortia. Genome Res. 22, 1813–1831 (2012).

    Article  CAS  Google Scholar 

  44. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  Google Scholar 

  45. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  Google Scholar 

  46. Crooks, G.E., Hon, G., Chandonia, J.M. & Brenner, S.E. WebLogo: a sequence logo generator. Genome Res. 14, 1188–1190 (2004).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S. Boswell for help with RNA-seq experiments and X. Zhang (Broad Institute) for help with p53 ChIP experiments, M. Springer for helpful discussions and advice on modeling, and members of the Lahav lab for comments and discussion. This research was supported by Novartis Institutes for Biomedical Research and NIH grant GM083303. A.H. was supported by Boehringer Ingelheim Fonds, PhD fellowship. J.S.-O. was supported by NIH grant CA207727. J.E.P. was supported by NIH grant GM102372. M.L.B. was supported by NIH grant HG003985.

Author information

Authors and Affiliations

Authors

Contributions

A.H., J.E.P., W.C.F. and G.L. conceived experiments. A.H., M.L.B. and G.L. conceived analyses. A.H. performed experiments and A.H. and J.S.-O. performed analyses. A.H. and G.L. wrote the paper.

Corresponding author

Correspondence to Galit Lahav.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Characterization of gene expression clusters shows p53 dependence and enrichment of known p53 target genes among induced genes. (related to Figure 1)

A. Quantiles (median in black, shaded area represents the 25% and 75% quantiles) of RNA-Seq data by cluster for MCF7 p53 wild-type cell line (blue) and MCF7 p53-shRNA cell line (gray). B. Mapping of the genes in each cluster to known target genes, obtained from Riley et al. 200830. P-values were calculated using the Binomial statistic, * = p-value<0.01. C. Top five enriched GO Biological Process categories in each cluster (FDR<0.05). GO Enrichment analysis was done using Enrichr software39,40. Both p-value and FDR are shown.

Supplementary Figure 2 Properties of p53 ChIP peaks that map to the induced clusters (related to Figure 3).

Distribution of maximal reads of p53 peaks per cluster. T-test was done to evaluate significance; ns stands for not significant (pval >0.2 for all comparisons). Black lines indicate the median, and the boxes and whiskers extend to the 25%-75% and the 5%-95% quantiles, respectively.

Supplementary Figure 3 Evaluation of model fit and parameters. (Related to Figures 4 and 5).

A. Distribution of R2 values for the model fit. Comparison between induced genes with p53 ChIP peak and induced genes without a p53 ChIP peak. B. Correlation of model derived kp values with maximal p53 ChIP peak signal for each gene. The signal from the closest peak was used for genes with multiple p53 ChIP peaks. C. Correlation of model derived kd values with published mRNA half-life data in MCF7 cells34. D. Schematic of gene expression dynamical features that were computed for the model stimulation. E-H. Simulation of the model (equation 1) for the shown range of kp and kd values. Heat maps of the gene expression properties derived for each kp and kd combination. I. Distribution of R2 values for the model fit for sustained data. J. Model simulation with p53 sustained input and only varying the kd parameter values.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–3 and Supplementary Table 1. (PDF 501 kb)

Life Sciences Reporting Summary (PDF 204 kb)

Supplementary Data Set 1

229 differentially expressed genes (Figure 1). (XLSX 67 kb)

Mean FPKM, log2(fold change) across 2 independent experiments as well as cluster assignments are provided.

Supplementary Data Set 2

p53 and H3K27ac ChIP peaks (Figure 2). (XLSX 4738 kb)

Genomic positions, normalized read counts per time-point for the ChIP and corresponding Input samples are provided. Closest gene assignments, with distance to TSS (bp) are provided for p53 ChIP peaks. Distance (bp) to the closest p53 ChIP peak is given for all H3K27ac peaks (used for Figure 2H).

Supplementary Data Set 3

p53 bound and differentially induced genes (Figure 3B) and results of model fit (Figure 4). (XLSX 11 kb)

Cluster number assignments and the kp and kd parameters resulting from the model fit (Figure 4) are provided.

Supplementary Data Set 4

Gene expression under the sustained p53 condition (Figure 5). (XLSX 16 kb)

Cluster assignments, mean FPKM values across 2 independent replicates and the resulting R2 values from the model prediction are provided.

Supplementary Data Set 5

Un-cropped western blot images for Figures 1A, 2A, 5A. (PDF 785 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hafner, A., Stewart-Ornstein, J., Purvis, J. et al. p53 pulses lead to distinct patterns of gene expression albeit similar DNA-binding dynamics. Nat Struct Mol Biol 24, 840–847 (2017). https://doi.org/10.1038/nsmb.3452

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.3452

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